Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.452
Filter
1.
Front Immunol ; 15: 1444045, 2024.
Article in English | MEDLINE | ID: mdl-39229279

ABSTRACT

Introduction: Colitis is an inflammatory bowel disease (IBD) characterized by immune cell dysregulation and alterations in the gut microbiome. In our previous report, we showed a natural product in cruciferous vegetables and ligand of the aryl hydrocarbon receptor (AhR), indole-3-carbinol (I3C), was able to reduce colitis-induced disease severity and microbial dysbiosis in an interleukin-22 (IL-22) dependent manner. Methods: In the current study, we performed single-cell RNA sequencing (scRNAseq) from colonocytes during colitis induction and supplementation with I3C and show how this treatment alters expression of genes involved in IL-22 signaling. To further define the role of IL-22 signaling in I3C-mediated protection during colitis and disease-associated microbial dysbiosis, we generated mice with AhR deficiency in RAR-related orphan receptor c (Rorc)-expressing cells (AhR ΔRorc ) which depletes this receptor in immune cells involved in production of IL-22. Colitis was induced in wildtype (WT), AhR ΔRorc , and littermate (LM) mice with or without I3C treatment. Results: Results showed AhR ΔRorc mice lost the efficacy effects of I3C treatment which correlated with a loss of ability to increase IL-22 by innate lymphoid type 3 (ILC3s), not T helper 22 (Th22) cells. 16S rRNA microbiome profiling studies showed AhR ΔRorc mice were unable to regulate disease-associated increases in Bacteroides, which differed between males and females. Lastly, inoculation with a specific disease-associated Bacteroides species, Bacteroides acidifaciens (B. acidifaciens), was shown to exacerbate colitis in females, but not males. Discussion: Collectively, this report highlights the cell and sex-specific role of AhR in regulating microbes that can impact colitis disease.


Subject(s)
Bacteroides , Colitis , Interleukin-22 , Interleukins , Receptors, Aryl Hydrocarbon , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Animals , Interleukins/metabolism , Colitis/immunology , Colitis/microbiology , Female , Mice , Male , Bacteroides/immunology , Gastrointestinal Microbiome/immunology , Dysbiosis/immunology , Mice, Inbred C57BL , Indoles/pharmacology , Disease Models, Animal , Sex Factors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Mice, Knockout
2.
Front Immunol ; 15: 1447021, 2024.
Article in English | MEDLINE | ID: mdl-39247203

ABSTRACT

Immune checkpoint inhibitors (ICIs) reinvigorate anti-tumor immune responses by disrupting co-inhibitory immune checkpoint molecules such as programmed cell death 1 (PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA-4). Although ICIs have had unprecedented success and have become the standard of care for many cancers, they are often accompanied by off-target inflammation that can occur in any organ system. These immune related adverse events (irAEs) often require steroid use and/or cessation of ICI therapy, which can both lead to cancer progression. Although irAEs are common, the detailed molecular and immune mechanisms underlying their development are still elusive. To further our understanding of irAEs and develop effective treatment options, there is pressing need for preclinical models recapitulating the clinical settings. In this review, we describe current preclinical models and immune implications of ICI-induced skin toxicities, colitis, neurological and endocrine toxicities, pneumonitis, arthritis, and myocarditis along with their management.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Humans , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/therapeutic use , Animals , Neoplasms/drug therapy , Neoplasms/immunology , Colitis/chemically induced , Colitis/immunology , Drug-Related Side Effects and Adverse Reactions , Immunotherapy/adverse effects , Immunotherapy/methods
3.
J Agric Food Chem ; 72(35): 19366-19377, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39178327

ABSTRACT

Proanthocyanidins (PA) have been proven to have an anti-inflammation effect in multiple models by regulating oxidative stress. ß-glucan (BG) could alleviate colitis from the perspectives of intestinal permeability and gut microbiota. In the present study, the synergistic anti-inflammatory function of PA and BG was explored from multiple aspects including immune response, intestinal barrier, gut microbiota, and differential metabolites. The results showed that the supplementation of PA and BG improved the colitis symptoms including atrophy of the colon, body weight loss, and organ index increase. Additionally, inflammatory cytokine levels and oxidative stress status were significantly regulated with the intake of PA and BG. Moreover, PA and BG intervention improved intestinal permeability and promoted the expression of barrier proteins. The microbiome and metabolic profile of cecal contents showed that PA and BG supplementation increased the abundance of anti-inflammatory bacteria and decreased the abundance of pro-inflammatory bacteria. Furthermore, some beneficial metabolites involved in amino acid metabolism, carbohydrate metabolism, and biosynthesis of other secondary metabolite pathways were increased. Overall, these findings have demonstrated the regulation of the inflammatory response and remodel of metabolite profiles by PA and BG complexes, indicating that it may serve as a new strategy for inflammatory bowel disease treatment in the future.


Subject(s)
Colitis , Dextran Sulfate , Gastrointestinal Microbiome , Mice, Inbred C57BL , Proanthocyanidins , beta-Glucans , Animals , beta-Glucans/administration & dosage , beta-Glucans/pharmacology , Dextran Sulfate/adverse effects , Mice , Proanthocyanidins/administration & dosage , Proanthocyanidins/pharmacology , Gastrointestinal Microbiome/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis/immunology , Male , Humans , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Bacteria/drug effects , Bacteria/metabolism , Anti-Inflammatory Agents/administration & dosage , Drug Synergism , Disease Models, Animal , Colon/metabolism , Colon/drug effects , Colon/immunology , Colon/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Oxidative Stress/drug effects
4.
Redox Biol ; 75: 103291, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39116526

ABSTRACT

Modulation of immune microenvironment is critical for inflammatory bowel disease (IBD) intervention. Epigallocatechin gallate (EGCG), as a natural low toxicity product, has shown promise in treating IBD. However, whether and how EGCG regulates the intestinal microenvironment is not fully understood. Here we report that EGCG lessens colitis by orchestrating Th1 polarization and self-amplification in a novel manner that required multilevel-regulated intestinal microecosystem. Mechanistically, EGCG activates GPR43 on IEC to inhibit Th1 polarization dependently of short chain fatty acid (SCFA)-producing gut microbiota. Inhibition of GPR43 activity weakens the protective effects of EGCG on colitis development. Moreover, we confirm that fecal SCFAs and/or intestinal GPR43 are limited in patients with colitis and are correlated with Th1 cell number. Taken together, our study reveals an intestinal microenvironment-dependent immunoregulatory effects of EGCG in treating IBD and provides insight into mechanisms of EGCG-based novel immunotherapeutic strategies for IBD.


Subject(s)
Catechin , Colitis , Gastrointestinal Microbiome , Receptors, G-Protein-Coupled , Th1 Cells , Catechin/analogs & derivatives , Catechin/pharmacology , Animals , Gastrointestinal Microbiome/drug effects , Mice , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Colitis/metabolism , Colitis/drug therapy , Colitis/immunology , Th1 Cells/immunology , Th1 Cells/drug effects , Th1 Cells/metabolism , Humans , Disease Models, Animal , Fatty Acids, Volatile/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology
5.
Sci Rep ; 14(1): 18618, 2024 08 10.
Article in English | MEDLINE | ID: mdl-39127850

ABSTRACT

This study aimed to investigate whether class A1 scavenger receptor (SR-A1) regulated macrophage polarization and gut microbial alteration during intestinal inflammation of colitis. A murine colitis model was established by feeding with dextran sulfate sodium (DSS), and treatment groups were injected intravenously with SR-A1 antibody. Results showed a preventive effect on colitis symptoms and fewer inflammatory cell infiltrates in treatment groups. Down-regulation of inflammatory cytokines and up-regulation of anti-inflammatory cytokine related to macrophages were seen in murine PBMC and LPMC after injected with SR-A1 antibody. The percentage of M2 macrophages was also elevated in treatment groups. In addition, SR-A1 antibody treatment resulted in the decreased apoptosis and increased proliferation of colonic epithelial cells. Other findings indicated that SR-A1 antibody injection could mediate its anti-inflammatory effect via inhibiting TLR4-MyD88-NF-kB signaling pathway and alterating the gut microbiota composition. Our research identified SR-A1 as a potential therapeutic target in inflammatory bowel disease (IBD).


Subject(s)
Colitis , Gastrointestinal Microbiome , Macrophages , Scavenger Receptors, Class A , Animals , Gastrointestinal Microbiome/drug effects , Colitis/immunology , Colitis/chemically induced , Colitis/microbiology , Colitis/metabolism , Mice , Macrophages/immunology , Macrophages/metabolism , Scavenger Receptors, Class A/metabolism , Dextran Sulfate/toxicity , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/immunology , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Disease Models, Animal , Cytokines/metabolism , Antibodies , NF-kappa B/metabolism , Mice, Inbred C57BL , Male , Apoptosis/drug effects
6.
Mol Immunol ; 173: 88-98, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39088935

ABSTRACT

Tanshinone I (Tan I) has been proven to exert an anti-inflammatory effect, but the complete mechanism remains unclear. In this study, Tan I was described to have no effect on Syk expression in resting or LPS-stimulated macrophages ex vivo, but dramatically suppressed Syk phosphorylation and CD80, CD86, and IL-1ß expression of macrophages. The inflammatory activity of macrophages in ApoC3-transgenic (ApoC3TG) mice is upregulated by Syk activation. Tan I was determined to downregulate Syk phosphorylation and inflammatory activity of macrophages in ApoC3TG mice, both ex vivo and in vivo. Intraperitoneal injection of Tan I (4 mg/kg) effectively alleviated DSS-induced colitis in mice, accompanying with suppressing the activation of intestinal macrophages. Mechanistically, Tan I-treated macrophages exhibited a decrease in cytoplasmic ROS, NLRP3, GSDMD, and IL-1ß, which suggested that the alternative pathway of inflammasome activation in macrophages was suppressed. The SPR assay demonstrated that Tan I bound to Syk protein with a dissociation constant (KD) of 2.473 × 10-6 M. When Syk expression was knocked down by its shRNA, the inhibitory effects of Tan I on macrophages were blocked. Collectively, Tanshinone I effectively alleviated DSS-induced colitis in mice by inhibiting Syk-stimulated inflammasome activation, hence suppressing the inflammatory activity of macrophages.


Subject(s)
Abietanes , Colitis , Dextran Sulfate , Inflammasomes , Macrophages , Syk Kinase , Animals , Syk Kinase/metabolism , Abietanes/pharmacology , Inflammasomes/metabolism , Inflammasomes/drug effects , Mice , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Colitis/chemically induced , Colitis/immunology , Colitis/drug therapy , Colitis/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Male
7.
J Immunol ; 213(6): 886-897, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39101764

ABSTRACT

Intestinal microbiota and selected strains of commensal bacteria influence regulatory T (Treg) cell functionality in the colon. Nevertheless, whether and how microbiota changes the transcriptome profile and TCR specificities of colonic Tregs remain to be precisely defined. In this study, we have employed single-cell RNA sequencing and comparatively analyzed colonic Tregs from specific pathogen-free and germ-free (GF) mice. We found that microbiota shifts the activation trajectory of colonic Tregs toward a distinct phenotypic subset enriched in specific pathogen-free but not in GF mice. Moreover, microbiota induced the expansion of specific Treg clonotypes with shared transcriptional specificities. The microbiota-induced subset of colonic Tregs, identified as PD-1- CXCR3+ Tregs, displayed enhanced suppressive capabilities compared with colonic Tregs derived from GF mice, enhanced production of IL-10, and were the primary regulators of enteric inflammation in dextran sodium sulfate-induced colitis. These findings identify a hitherto unknown gut microbiota and immune cell interaction module that could contribute to the development of a therapeutic modality for intestinal inflammatory diseases.


Subject(s)
Colitis , Colon , Gastrointestinal Microbiome , Receptors, Antigen, T-Cell , T-Lymphocytes, Regulatory , Animals , Gastrointestinal Microbiome/immunology , Mice , T-Lymphocytes, Regulatory/immunology , Colon/immunology , Colon/microbiology , Colitis/immunology , Receptors, Antigen, T-Cell/immunology , Mice, Inbred C57BL , Dextran Sulfate , Specific Pathogen-Free Organisms , Interleukin-10/immunology
8.
Int Immunopharmacol ; 140: 112884, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39133959

ABSTRACT

Multiple lines of evidence suggest that Retinoic Acid Related Orphan Nuclear Receptor gamma t (RORγt) is a potent therapeutic target for inflammatory bowel disease (IBD). However, systemic blockade of RORγt easily leads to thymic lymphoma and aberrant liver function. Therefore, the development of gut-limited RORγt antagonists may lead to the development of innovative IBD therapeutics that improve safety and retain effectiveness. We discovered SPH7854, a potent and selective RORγt antagonist. The effect of SPH7854 on the differentiation of T helper 1 (Th1)/Th17/regulatory T (Treg) cells was evaluated in mouse and human primary cells. SPH7854 (2-(4-(ethylsulfonyl)phenyl)-N- (6-(2-methyl-2-(pyridin-2-yl) propanoyl)pyridin-3-yl)acetamide) dose-dependently inhibited interleukin-17A (IL-17A) secretion from mouse CD4 + T cells and human peripheral blood mononuclear cells (PBMC). Additionally, SPH7854 strongly suppressed Th17 cell differentiation and considerably promoted Treg cell differentiation while slightly affected Th1 cell differentiation from mouse CD4 + T cells. The pharmacokinetic (PK) studies indicated that SPH7854 was restricted to the gut: the bioavailability and maximal plasma concentration of SPH7854 after oral administration (6 mg/kg) were 1.24 ± 0.33 % and 4.92 ± 11.81 nM, respectively, in rats. Strikingly, oral administration of SPH7854 (5 mg/kg and 15 mg/kg) twice daily significantly alleviated 2, 4, 6-trinitrobenzensulfonic acid (TNBS)-induced colitis in rats. SPH7854, especially at 15 mg/kg, significantly alleviated symptoms and improved macroscopic signs and microscopic structure in rat colitis, with decreased colonic mucosal levels of IL-17A, IL-6, tumor necrosis factor α (TNFα), monocyte chemoattractant protein-1 (MCP-1) and myeloperoxidase (MPO). These evidences indicated that blockade of RORγt activity via a gut-limited antagonist may be an effective and safe therapeutic strategy for IBD treatment.


Subject(s)
Colitis , Nuclear Receptor Subfamily 1, Group F, Member 3 , Th17 Cells , Trinitrobenzenesulfonic Acid , Animals , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Humans , Colitis/chemically induced , Colitis/drug therapy , Colitis/immunology , Male , Rats , Mice , Th17 Cells/immunology , Th17 Cells/drug effects , Rats, Sprague-Dawley , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Cell Differentiation/drug effects , Disease Models, Animal , Interleukin-17/metabolism , Interleukin-17/antagonists & inhibitors , Acetamides/therapeutic use , Acetamides/pharmacology , Cells, Cultured , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Colon/drug effects , Colon/pathology , Colon/immunology , Mice, Inbred C57BL
9.
Nat Immunol ; 25(9): 1565-1579, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39103576

ABSTRACT

Nucleophosmin 1 (NPM1) is commonly mutated in myelodysplastic syndrome (MDS) and acute myeloid leukemia. Concurrent inflammatory bowel diseases (IBD) and MDS are common, indicating a close relationship between IBD and MDS. Here we examined the function of NPM1 in IBD and colitis-associated colorectal cancer (CAC). NPM1 expression was reduced in patients with IBD. Npm1+/- mice were more susceptible to acute colitis and experimentally induced CAC than littermate controls. Npm1 deficiency impaired the function of interleukin-22 (IL-22)-producing group three innate lymphoid cells (ILC3s). Mice lacking Npm1 in ILC3s exhibited decreased IL-22 production and accelerated development of colitis. NPM1 was important for mitochondrial biogenesis and metabolism by oxidative phosphorylation in ILC3s. Further experiments revealed that NPM1 cooperates with p65 to promote mitochondrial transcription factor A (TFAM) transcription in ILC3s. Overexpression of Npm1 in mice enhanced ILC3 function and reduced the severity of dextran sulfate sodium-induced colitis. Thus, our findings indicate that NPM1 in ILC3s protects against IBD by regulating mitochondrial metabolism through a p65-TFAM axis.


Subject(s)
Colitis , Immunity, Mucosal , Mice, Knockout , Mitochondria , Nuclear Proteins , Nucleophosmin , Oxidative Phosphorylation , Animals , Mitochondria/metabolism , Mice , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Humans , Colitis/immunology , Colitis/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred C57BL , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Interleukin-22 , Immunity, Innate , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Dextran Sulfate , Male , Interleukins/metabolism , Interleukins/genetics , Interleukins/immunology , Female
10.
BMJ Case Rep ; 17(7)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39038878

ABSTRACT

Anticancer immunotherapies modulate the body's immune system to recognise and eradicate cancerous cells. However, stimulation of the body's immune system can also lead to a number of adverse effects when those immune cells target non-cancerous cells in the form of autoimmunity. One relatively common example of this off-target action is colitis.We present three patients who presented atypically with colitis, consequently, leading to a delayed diagnosis. These cases highlight the diverse ways a relatively common immune-related adverse event can present.


Subject(s)
Colitis , Constipation , Humans , Constipation/etiology , Colitis/immunology , Male , Female , Middle Aged , Aged , Delayed Diagnosis
11.
Cell Mol Life Sci ; 81(1): 327, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39085655

ABSTRACT

Dysregulation of mucosal immune system has been proposed to be critical in the pathogenesis of inflammatory bowel diseases (IBDs). Regulatory T cells (Tregs) play an important role in regulating immune responses. Tregs are involved in maintaining intestinal homeostasis and exerting suppressive function in colitis. Our previous studies showed that a novel forkhead box protein P3 (Foxp3) negative Tregs (Treg-of-B cells), induced by culturing naïve CD4+ T cells with B cells, could protect against colitis and downregulate T helper (Th) 1 and Th17 cell cytokines in T cell-mediated colitis. In the present study, we aimed to induce Treg-of-B cells in the CD8+ T-cell population and investigate their characteristics and immunomodulatory functions. Our results showed that CD8+ Treg-of-B cells expressed Treg-associated markers, including lymphocyte-activation gene-3 (LAG3), inducible co-stimulator (ICOS), programmed death-1 (PD-1), cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), tumor necrosis factor receptor superfamily member-4 (TNFRSF4, OX40), and tumor necrosis factor receptor superfamily member-18 (TNFRSF18, GITR), but did not express Foxp3. CD8+ Treg-of-B cells produced higher concentration of inhibitory cytokine interleukin (IL)-10, and expressed higher levels of cytotoxic factor granzyme B and perforin after stimulation, compared to those of CD8+CD25- T cells. Moreover, CD8+ Treg-of-B cells suppressed T cell proliferation in vitro and alleviated colonic inflammation in chronic dextran sulfate sodium (DSS)-induced colitis. In conclusion, our study identified a novel subpopulation of CD8+ Tregs with suppressive effects through cell contact. These CD8+ Treg-of-B cells might have therapeutic potential for IBDs.


Subject(s)
CD8-Positive T-Lymphocytes , Disease Models, Animal , Inflammatory Bowel Diseases , Mice, Inbred C57BL , T-Lymphocytes, Regulatory , Animals , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Colitis/immunology , Colitis/pathology , Colitis/chemically induced , Dextran Sulfate , Forkhead Transcription Factors/metabolism , Interleukin-10/metabolism , Interleukin-10/immunology
12.
Cell Rep ; 43(7): 114458, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38996070

ABSTRACT

Regulatory T (Treg) cells play a critical regulatory role in the immune system by suppressing excessive immune responses and maintaining immune balance. The effective migration of Treg cells is crucial for controlling the development and progression of inflammatory diseases. However, the mechanisms responsible for directing Treg cells into the inflammatory tissue remain incompletely elucidated. In this study, we identified BAF60b, a subunit of switch/sucrose nonfermentable (SWI/SNF) chromatin remodeling complexes, as a positive regulator of Treg cell migration that inhibits the progression of inflammation in experimental autoimmune encephalomyelitis (EAE) and colitis animal models. Mechanistically, transcriptome and genome-wide chromatin-landscaped analyses demonstrated that BAF60b interacts with the transcription factor RUNX1 to promote the expression of CCR9 on Treg cells, which in turn affects their ability to migrate to inflammatory tissues. Our work provides insights into the essential role of BAF60b in regulating Treg cell migration and its impact on inflammatory diseases.


Subject(s)
Cell Movement , Inflammation , Mice, Inbred C57BL , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Mice , Inflammation/pathology , Inflammation/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Humans , Transcription Factors/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Colitis/metabolism , Colitis/pathology , Colitis/immunology , Colitis/genetics
13.
Curr Protoc ; 4(7): e1092, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39007482

ABSTRACT

The intestinal inflammation induced by injection of naïve CD4+ T cells into lymphocyte-deficient hosts (more commonly known as the T cell transfer model of colitis) shares many features of idiopathic inflammatory bowel disease (IBD) in humans, such as epithelial cell hyperplasia, crypt abscess formation, and dense lamina propria lymphocyte infiltration. As such, it provides a useful tool for studying mucosal immune regulation as it relates to the pathogenesis and treatment of IBD in humans. In the IBD model described here, colitis is induced in Rag (recombination-activating gene)-deficient mice by reconstitution of these mice with naïve CD4+CD45RBhi T cells through adoptive T cell transfer. Although different recipient hosts of cell transfer can be used, Rag-deficient mice are the best characterized and support studies that are both flexible and reproduceable. As described in the Basic Protocol, in most studies the transferred cells consist of naïve CD4+ T cells (CD45RBhi T cells) derived by fluorescence-activated cell sorting from total CD4+ T cells previously purified using immunomagnetic negative selection beads. In a Support Protocol, methods to characterize colonic disease progression are described, including the monitoring of weight loss and diarrhea and the histological assessment of colon pathology. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Induction of IBD in Rag-deficient mice by the transfer of naïve CD4+CD45RBhi T cells Support Protocol: Monitoring development of colitis.


Subject(s)
CD4-Positive T-Lymphocytes , Inflammatory Bowel Diseases , Animals , Mice , Adoptive Transfer , CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Colitis/chemically induced , Colitis/pathology , Disease Models, Animal , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology
14.
Biomolecules ; 14(7)2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39062588

ABSTRACT

Guanylate binding protein 5 (GBP5) is an emerging immune component that has been increasingly recognized for its involvement in autoimmune diseases, particularly inflammatory bowel disease (IBD). IBD is a complex disease involving inflammation of the gastrointestinal tract. Here, we explored the functional significance of GBP5 using Gbp5 knockout mice and wildtype mice exposed to dextran sulfate sodium (DSS) to generate chronic colitis model. We found that Gbp5 deficiency protected mice from DSS-induced chronic colitis. Transcriptome analysis of colon tissues showed reduced immune responses in Gbp5 knockout mice compared to those in corresponding wildtype mice. We further observed that after repeated DSS exposure, the gut microbiota was altered, both in wildtype mice and Gbp5 knockout mice; however, the gut microbiome health index was higher in the Gbp5 knockout mice. Notably, a probiotic murine commensal bacterium, Dubosiella, was predominantly enriched in these knockout mice. Our findings suggest that GBP5 plays an important role in promoting inflammation and dysbiosis in the intestine, the prevention of which might therefore be worth exploring in regards to IBD treatment.


Subject(s)
Colitis , Dextran Sulfate , Disease Models, Animal , Gastrointestinal Microbiome , Mice, Knockout , Animals , Mice , Chronic Disease , Colitis/microbiology , Colitis/chemically induced , Colitis/immunology , Colitis/genetics , Colitis/metabolism , Dysbiosis/microbiology , Dysbiosis/immunology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/deficiency , Mice, Inbred C57BL
15.
J Immunol ; 213(4): 481-493, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38949555

ABSTRACT

Aberrant activity of NLRP3 has been shown associations with severe diseases. Palmitoylation is a kind of protein post-translational modification, which has been shown to regulate cancer development and the innate immune system. Here, we showed that NLRP3 is palmitoylated at Cys419 and that palmitoyltransferase ZDHHC17 is the predominant enzyme that mediates NLRP3 palmitoylation and promotes NLRP3 activation by interacting with NLRP3 and facilitating NIMA-related kinase 7 (NEK7)-NLRP3 interactions. Blockade of NLRP3 palmitoylation by a palmitoylation inhibitor, 2-bromopalmitate, effectively inhibited NLRP3 activation in vitro. Also, in a dextran sulfate sodium-induced colitis model in mice, 2-bromopalmitate application could attenuate weight loss, improve the survival rate, and rescue pathological changes in the colon of mice. Overall, our study reveals that palmitoylation of NLPR3 modulates inflammasome activation and inflammatory bowel disease development. We propose that drugs targeting NLRP3 palmitoylation could be promising candidates in the treatment of NLRP3-mediated inflammatory diseases.


Subject(s)
Acyltransferases , Colitis , Inflammasomes , Inflammatory Bowel Diseases , Lipoylation , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Mice , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Humans , Acyltransferases/metabolism , Colitis/immunology , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate/toxicity , Mice, Inbred C57BL , NIMA-Related Kinases/metabolism , Palmitates/pharmacology , Disease Models, Animal , HEK293 Cells , Protein Processing, Post-Translational
16.
Int J Biol Macromol ; 276(Pt 1): 133794, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38992530

ABSTRACT

Inflammatory Bowel Disease (IBD) is a chronic condition whose incidence has been rising globally. Synbiotic (SYN) is an effective means of preventing IBD. This study investigated the preventive effects and potential biological mechanisms of SYN (Bifidobacterium longum, Lactobacillus acidophilus, and sea buckthorn polysaccharides) on DSS-induced colitis in mice. The results indicated that dietary supplementation with SYN has a significant improvement effect on DSS mice. SYN ameliorated disease activity index (DAI), colon length, and intestinal barrier permeability in mice. In addition, RT-qPCR results indicated that after SYN intervention, the expression levels of pro-inflammatory factors (IL-6, IL-1ß, TNF-α, and IL-17F) and transcription factor RORγt secreted by Th17 cells were significantly reduced, and the expression levels of anti-inflammatory factors (IL-10 and TGF-ß) and transcription factor Foxp3 secreted by Treg cells were robustly increased. 16S rDNA sequencing analysis revealed that key intestinal microbiota related to Th17/Treg balance (Ligilactobacillus, Lactobacillus, Bacteroides, and Akkermansia) was significantly enriched. At the same time, a significant increase in microbial metabolites SCFAs and BAs was observed. We speculate that SYN may regulate the Th17/Treg balance by restructuring the structure and composition of the intestinal microbiota, thereby mitigating DSS-induced colitis.


Subject(s)
Colitis , Dextran Sulfate , Gastrointestinal Microbiome , Hippophae , Polysaccharides , Synbiotics , T-Lymphocytes, Regulatory , Th17 Cells , Animals , Gastrointestinal Microbiome/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism , Colitis/chemically induced , Colitis/immunology , Colitis/metabolism , Mice , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Polysaccharides/pharmacology , Hippophae/chemistry , Fatty Acids, Volatile/metabolism , Homeostasis/drug effects , Male , Cytokines/metabolism , Disease Models, Animal , Mice, Inbred C57BL
17.
Inflamm Res ; 73(9): 1493-1510, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38981913

ABSTRACT

OBJECTIVE AND DESIGN: Compelling evidence indicates that dysregulated macrophages may play a key role in driving inflammation in inflammatory bowel disease (IBD). Fibroblast growth factor (FGF)-19, which is secreted by ileal enterocytes in response to bile acids, has been found to be significantly lower in IBD patients compared to healthy individuals, and is negatively correlated with the severity of diarrhea. This study aims to explore the potential impact of FGF19 signaling on macrophage polarization and its involvement in the pathogenesis of IBD. METHODS: The dextran sulfate sodium (DSS)-induced mouse colitis model was utilized to replicate the pathology of human IBD. Mice were created with a conditional knockout of FGFR4 (a specific receptor of FGF19) in myeloid cells, as well as mice that overexpressing FGF19 specifically in the liver. The severity of colitis was measured using the disease activity index (DAI) and histopathological staining. Various techniques such as Western Blotting, quantitative PCR, flow cytometry, and ELISA were employed to assess polarization and the expression of inflammatory genes. RESULTS: Myeloid-specific FGFR4 deficiency exacerbated colitis in the DSS mouse model. Deletion or inhibition of FGFR4 in bone marrow-derived macrophages (BMDMs) skewed macrophages towards M1 polarization. Analysis of transcriptome sequencing data revealed that FGFR4 deletion in macrophages significantly increased the activity of the complement pathway, leading to an enhanced inflammatory response triggered by LPS. Mechanistically, FGFR4-knockout in macrophages promoted complement activation and inflammatory response by upregulating the nuclear factor-κB (NF-κB)-pentraxin3 (PTX3) pathway. Additionally, FGF19 suppressed these pathways and reduced inflammatory response by activating FGFR4 in inflammatory macrophages. Liver-specific overexpression of FGF19 also mitigated inflammatory responses induced by DSS in vivo. CONCLUSION: Our study highlights the significance of FGF19-FGFR4 signaling in macrophage polarization and the pathogenesis of IBD, offering a potential new therapeutic target for IBD.


Subject(s)
Colitis , Dextran Sulfate , Fibroblast Growth Factors , Macrophages , Receptor, Fibroblast Growth Factor, Type 4 , Animals , Male , Mice , Colitis/chemically induced , Colitis/pathology , Colitis/immunology , Colon/pathology , Colon/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Liver/pathology , Liver/metabolism , Macrophages/metabolism , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism
18.
Commun Biol ; 7(1): 914, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075283

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) play key roles in intestinal inflammation. Olfactomedin 4 (OLFM4) is highly expressed in the colon and has a potential role in dextran sodium sulfate-induced colitis. However, the detailed mechanisms underlying the effects of OLFM4 on ILC3-mediated colitis remain unclear. In this study, we identify OLFM4 as a positive regulator of IL-22+ILC3. OLFM4 expression in colonic ILC3s increases substantially during intestinal inflammation in humans and mice. Compared to littermate controls, OLFM4-deficient (OLFM4-/-) mice are more susceptible to bacterial infection and display greater resistance to anti-CD40 induced innate colitis, together with impaired IL-22 production by ILC3, and ILC3s from OLFM4-/-mice are defective in pathogen resistance. Besides, mice with OLFM4 deficiency in the RORγt compartment exhibit the same trend as in OLFM4-/-mice, including colonic inflammation and IL-22 production. Mechanistically, the decrease in IL-22+ILC3 caused by OLFM4 deficiency involves the apoptosis signal-regulating kinase 1 (ASK1)- p38 MAPK signaling-dependent downregulation of RAR-related orphan receptor gamma (RORγt) protein. The OLFM4-metadherin (MTDH) complex upregulates p38/RORγt signaling, which is necessary for IL-22+ILC3 activation. The findings indicate that OLFM4 is a novel regulator of IL-22+ILC3 and essential for modulating intestinal inflammation and tissue homeostasis.


Subject(s)
Colitis , Interleukin-22 , Interleukins , Mice, Knockout , Animals , Mice , Interleukins/metabolism , Interleukins/genetics , Colitis/genetics , Colitis/chemically induced , Colitis/metabolism , Colitis/immunology , Colitis/pathology , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred C57BL , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/genetics , Immunity, Innate , Inflammation/metabolism , Inflammation/genetics , Male , Glycoproteins
19.
Int Immunopharmacol ; 140: 112825, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39079347

ABSTRACT

BACKGROUND: Exosomes derived from endometrial regenerative cells (ERC-Exos) can inherit the immunomodulatory function from ERCs, however, whether ERC-Exos exhibit such effect on inflammatory bowel diseases with mucosal immune dysregulation has not been explored. Insulin-like growth factor-Ⅱ (IGF2) is considered to possess the potential to induce an anti-inflammatory phenotype in immune cells. In this study, the contribution of IGF2 in mediating the protective efficacy of ERC-Exos on colitis was investigated. METHODS: Lentiviral transfection was employed to obtain IGF2-specific knockout ERC-Exos (IGF2-/--ERC-Exos). Experimental colitis mice induced by dextran sulfate sodium (DSS) were divided into the phosphate-buffered saline (untreated), ERC-Exos-treated and IGF2-/--ERC-Exos-treated groups. Colonic histopathological analysis and intestinal barrier function were explored. The infiltration of CD4+ T cells and dendritic cells (DCs) were analyzed by immunofluorescence staining and flow cytometry. The maturation and function of bone marrow-derived dendritic cells (BMDCs) in different exosome administrations were evaluated by flow cytometry, ELISA and the coculture system, respectively. RESULTS: Compared with the untreated group, ERC-Exos treatment significantly attenuated DSS-induced weight loss, bloody stools, shortened colon length, pathological damage, as well as repaired the weakened intestinal mucosal barrier, including promoting the goblet cells retention, restoring the intestinal barrier integrity and enhancing the expression of tight junction proteins, while the protective effect of exosomes was impaired with the knockout of IGF2 in ERC-Exos. Additionally, IGF2-expressing ERC-Exos decreased the proportions of Th1 and Th17, increased the proportions of Treg, as well as attenuated DC infiltration and maturation in mesenteric lymph nodes and lamina propria of the colitis mice. ERC-Exos were also observed to be phagocytosed by BMDCs and IGF2 is responsible for the modulating effect of ERC-Exos on BMDCs in vitro. CONCLUSIONS: Exosomes derived from ERCs can exert a therapeutic effect on experimental colitis with remarkable alleviation of the intestinal barrier damage and the abnormal mucosal immune responses. We emphasized that IGF2 plays a critical role for ERC-Exos mediated immunomodulatory function and protection against colitis.


Subject(s)
Colitis , Dextran Sulfate , Endometrium , Exosomes , Insulin-Like Growth Factor II , Animals , Female , Humans , Mice , Cells, Cultured , Colitis/chemically induced , Colitis/immunology , Colitis/therapy , Colon/pathology , Colon/immunology , Dendritic Cells/immunology , Disease Models, Animal , Endometrium/immunology , Endometrium/pathology , Exosomes/metabolism , Exosomes/transplantation , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Mice, Knockout , Regeneration
20.
Front Immunol ; 15: 1401949, 2024.
Article in English | MEDLINE | ID: mdl-39076972

ABSTRACT

Introduction: Ubiquitin-specific proteases (USPs), a large subset of more than 50 deubiquitinase proteins, have recently emerged as promising targets in cancer. However, their role in immune cell regulation, particularly in T cell activation, differentiation, and effector functions, remains largely unexplored. Methods: We utilized a USP28 knockout mouse line to study the effect of USP28 on T cell activation and function, and its role in intestinal inflammation using the dextran sulfate sodium (DSS)-induced colitis model and a series of in vitro assays. Results: Our results show that USP28 exerts protective effects in acute intestinal inflammation. Mechanistically, USP28 knockout mice (USP28-/-) exhibited an increase in total T cells mainly due to an increased CD8+ T cell content. Additionally, USP28 deficiency resulted in early defects in T cell activation and functional changes. Specifically, we observed a reduced expression of IL17 and an increase in inducible regulatory T (iTreg) suppressive functions. Importantly, activated T cells lacking USP28 showed increased STAT5 phosphorylation. Consistent with these findings, these mice exhibited increased susceptibility to acute DSS-induced intestinal inflammation, accompanied by elevated IL22 cytokine levels. Conclusions: Our findings demonstrate that USP28 is essential for T cell functionality and protects mice from acute DSS-induced colitis by regulating STAT5 signaling and IL22 production. As a T cell regulator, USP28 plays a crucial role in immune responses and intestinal health.


Subject(s)
Colitis , Interleukin-22 , Interleukins , STAT5 Transcription Factor , Ubiquitin Thiolesterase , Animals , Mice , Colitis/chemically induced , Colitis/immunology , Colitis/metabolism , Dextran Sulfate , Disease Models, Animal , Inflammation/immunology , Inflammation/metabolism , Interleukins/metabolism , Interleukins/genetics , Intestines/immunology , Intestines/pathology , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , STAT5 Transcription Factor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL