Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.432
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Nutrients ; 16(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38892639

ABSTRACT

Compared to the general population, patients with inflammatory bowel disease (IBD) are less likely to be vaccinated, putting them at an increased risk of vaccine-preventable illnesses. This risk is further compounded by the immunosuppressive therapies commonly used in IBD management. Therefore, developing new treatments for IBD that maintain immune function is crucial, as successful management can lead to better vaccination outcomes and overall health for these patients. Here, we investigate the potential of recombinant banana lectin (rBanLec) as a supporting therapeutic measure to improve IBD control and possibly increase vaccination rates among IBD patients. By examining the therapeutic efficacy of rBanLec in a murine model of experimental colitis, we aim to lay the foundation for its application in improving vaccination outcomes. After inducing experimental colitis in C57BL/6 and BALB/c mice with 2,4,6-trinitrobenzene sulfonic acid, we treated animals orally with varying doses of rBanLec 0.1-10 µg/mL (0.01-1 µg/dose) during the course of the disease. We assessed the severity of colitis and rBanLec's modulation of the immune response compared to control groups. rBanLec administration resulted in an inverse dose-response reduction in colitis severity (less pronounced weight loss, less shortening of the colon) and an improved recovery profile, highlighting its therapeutic potential. Notably, rBanLec-treated mice exhibited significant modulation of the immune response, favoring anti-inflammatory pathways (primarily reduction in a local [TNFα]/[IL-10]) crucial for effective vaccination. Our findings suggest that rBanLec could mitigate the adverse effects of immunosuppressive therapy on vaccine responsiveness in IBD patients. By improving the underlying immune response, rBanLec may increase the efficacy of vaccinations, offering a dual benefit of disease management and prevention of vaccine-preventable illnesses. Further studies are required to translate these findings into clinical practice.


Subject(s)
Colitis , Disease Models, Animal , Inflammatory Bowel Diseases , Mice, Inbred BALB C , Mice, Inbred C57BL , Musa , Animals , Inflammatory Bowel Diseases/drug therapy , Mice , Musa/chemistry , Colitis/drug therapy , Colitis/immunology , Colitis/prevention & control , Plant Lectins/pharmacology , Trinitrobenzenesulfonic Acid , Immunomodulating Agents/pharmacology , Female , Colon/drug effects , Colon/immunology , Colon/pathology , Male
9.
Sci Immunol ; 9(96): eadd6774, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38875317

ABSTRACT

Pro-inflammatory CD4+ T cells are major drivers of autoimmune diseases, yet therapies modulating T cell phenotypes to promote an anti-inflammatory state are lacking. Here, we identify T helper 17 (TH17) cell plasticity in the kidneys of patients with antineutrophil cytoplasmic antibody-associated glomerulonephritis on the basis of single-cell (sc) T cell receptor analysis and scRNA velocity. To uncover molecules driving T cell polarization and plasticity, we established an in vivo pooled scCRISPR droplet sequencing (iCROP-seq) screen and applied it to mouse models of glomerulonephritis and colitis. CRISPR-based gene targeting in TH17 cells could be ranked according to the resulting transcriptional perturbations, and polarization biases into T helper 1 (TH1) and regulatory T cells could be quantified. Furthermore, we show that iCROP-seq can facilitate the identification of therapeutic targets by efficient functional stratification of genes and pathways in a disease- and tissue-specific manner. These findings uncover TH17 to TH1 cell plasticity in the human kidney in the context of renal autoimmunity.


Subject(s)
Single-Cell Analysis , Th17 Cells , Animals , Humans , Mice , Th17 Cells/immunology , Glomerulonephritis/immunology , Glomerulonephritis/genetics , Cell Plasticity/immunology , Cell Plasticity/genetics , Kidney/immunology , Kidney/pathology , Mice, Inbred C57BL , CRISPR-Cas Systems , Colitis/immunology , Colitis/genetics , Inflammation/immunology , Inflammation/genetics , Female , Male , Clustered Regularly Interspaced Short Palindromic Repeats/immunology
10.
Gut Microbes ; 16(1): 2363020, 2024.
Article in English | MEDLINE | ID: mdl-38841892

ABSTRACT

CD4+ T cells play a critical role in regulating autoimmune diseases, and intestinal microbial metabolites control various immune responses. Granzyme B (GzmB)-producing CD4+ T cells have been recently reported to participate in the pathogenesis of autoimmune diseases. Here, we found that GzmbB-deficient CD4+ T cells induced more severe colitis in Rag1-/- mice than wild-type (WT) CD4+ T cells. Germ-free (GF) mice exhibited a lower expression of GzmB in intestinal CD4+ T cells compared to specific pathogen-free (SPF) mice. Intestinal microbial metabolite butyrate increased GzmB expression in CD4+ T cells, especially in IL-10-producing Th1 cells, through HDAC inhibition and GPR43, but not GPR41 and GPR109a. Butyrate-treated GzmB-deficient CD4+ T cells demonstrated more severe colitis compared to butyrate-treated WT CD4+ T cells in the T cell transfer model. Butyrate altered intestinal microbiota composition, but altered microbiota did not mediate butyrate induction of intestinal CD4+ T cell expression of GzmB in mice. Blimp1 was involved in the butyrate induction of GzmB in IL-10-producing Th1 cells. Glucose metabolism, including glycolysis and pyruvate oxidation, mediated butyrate induction of GzmB in Th1 cells. In addition, we found that IKZF3 and NR2F6 regulated GzmB expression induced by butyrate. Together, our studies underscored the critical role of GzmB in mediating gut bacterial metabolite butyrate regulation of T cell tolerance at the mucosal surface.


Subject(s)
Butyrates , Colitis , Gastrointestinal Microbiome , Granzymes , Interleukin-10 , Mice, Inbred C57BL , Th1 Cells , Animals , Interleukin-10/metabolism , Interleukin-10/genetics , Interleukin-10/immunology , Th1 Cells/immunology , Mice , Gastrointestinal Microbiome/drug effects , Butyrates/metabolism , Butyrates/pharmacology , Granzymes/metabolism , Colitis/immunology , Colitis/microbiology , Colitis/metabolism , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Immune Tolerance , Homeodomain Proteins
11.
Cell Immunol ; 401-402: 104839, 2024.
Article in English | MEDLINE | ID: mdl-38850753

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) is a chronic and relapsing disease characterized by immune-mediated dysfunction of intestinal homeostasis. Alteration of the enteric nervous system and the subsequent neuro-immune interaction are thought to contribute to the initiation and progression of IBD. However, the role of dopamine beta-hydroxylase (DBH), an enzyme converting dopamine into norepinephrine, in modulating intestinal inflammation is not well defined. METHODS: CD4+CD45RBhighT cell adoptive transfer, and 2,4-dinitrobenzene sulfonic acid (DNBS) or dextran sodium sulfate (DSS)-induced colitis were collectively conducted to uncover the effects of DBH inhibition by nepicastat, a DBH inhibitor, in mucosal ulceration, disease severity, and T cell function. RESULTS: Inhibition of DBH by nepicastat triggered therapeutic effects on T cell adoptive transfer induced chronic mouse colitis model, which was consistent with the gene expression of DBH in multiple cell populations including T cells. Furthermore, DBH inhibition dramatically ameliorated the disease activity and colon shortening in chemically induced acute and chronic IBD models, as evidenced by morphological and histological examinations. The reshaped systemic inflammatory status was largely associated with decreased pro-inflammatory mediators, such as TNF-α, IL-6 and IFN-γ in plasma and re-balanced Th1, Th17 and Tregs in mesenteric lymph nodes (MLNs) upon colitis progression. Additionally, the conversion from dopamine (DA) to norepinephrine (NE) was inhibited resulting in increase in DA level and decrease in NE level and DA/NE showed immune-modulatory effects on the activation of immune cells. CONCLUSION: Modulation of neurotransmitter levels via inhibition of DBH exerted protective effects on progression of murine colitis by modulating the neuro-immune axis. These findings suggested a promising new therapeutic strategy for attenuating intestinal inflammation.


Subject(s)
Adoptive Transfer , Colitis , Dopamine beta-Hydroxylase , Inflammatory Bowel Diseases , Lymphocyte Activation , Mice, Inbred C57BL , Animals , Mice , Colitis/chemically induced , Colitis/immunology , Dopamine beta-Hydroxylase/metabolism , Inflammatory Bowel Diseases/immunology , Lymphocyte Activation/immunology , Disease Models, Animal , Dextran Sulfate , Inflammation/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Male , Cytokines/metabolism
12.
Sci Immunol ; 9(96): eadi8954, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905325

ABSTRACT

Intestinal inflammation shifts microbiota composition and metabolism. How the host monitors and responds to such changes remains unclear. Here, we describe a protective mechanism by which mucosal-associated invariant T (MAIT) cells detect microbiota metabolites produced upon intestinal inflammation and promote tissue repair. At steady state, MAIT ligands derived from the riboflavin biosynthesis pathway were produced by aerotolerant bacteria residing in the colonic mucosa. Experimental colitis triggered luminal expansion of riboflavin-producing bacteria, leading to increased production of MAIT ligands. Modulation of intestinal oxygen levels suggested a role for oxygen in inducing MAIT ligand production. MAIT ligands produced in the colon rapidly crossed the intestinal barrier and activated MAIT cells, which expressed tissue-repair genes and produced barrier-promoting mediators during colitis. Mice lacking MAIT cells were more susceptible to colitis and colitis-driven colorectal cancer. Thus, MAIT cells are sensitive to a bacterial metabolic pathway indicative of intestinal inflammation.


Subject(s)
Colitis , Dysbiosis , Gastrointestinal Microbiome , Mice, Inbred C57BL , Mucosal-Associated Invariant T Cells , Animals , Mucosal-Associated Invariant T Cells/immunology , Colitis/immunology , Colitis/microbiology , Dysbiosis/immunology , Mice , Gastrointestinal Microbiome/immunology , Mice, Knockout , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Riboflavin/immunology
13.
Int Immunopharmacol ; 137: 112417, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38897122

ABSTRACT

Drug local delivery system that directly supply anti-cancer drugs to the tumor microenvironment (TME) results in excellent tumor control and minimizes side effects associated with the anti-cancer drugs. Immune checkpoint inhibitors (ICIs) have been the mainstay of cancer immunotherapy. However, the systemic administration of ICIs is accompanied by considerable immunotherapy-related toxicity. To explore whether an anti-PD-L1 antibody administered locally via a sustained-release gel-forming carrier retains its effective anticancer function while causing fewer colitis-like side effects, CT, a previously reported depot system, was used to locally deliver an anti-PD-L1 antibody together with curcumin to the TME in bladder cancer-bearing ulcerative colitis model mice. We showed that CT-mediated intratumoral coinjection of an anti-PD-L1 antibody and curcumin enabled sustained release of both the loaded anti-PD-L1 antibody and curcumin, which contributed to substantial anticancer effects with negligible side effects on the colons of the UC model mice. However, although the anti-PD-L1 antibody administered systemically synergized with the CT-mediated intratumoral delivery of curcumin in inhibiting tumour growth, colitis was significantly worsened by intraperitoneal administration of anti-PD-L1 antibody. These findings suggested that CT is a promising agent for the local delivery of anticancer drugs, as it can allow effective anticancer functions to be retained while sharply reducing the adverse side effects associated with the systemic administration of these drugs.


Subject(s)
B7-H1 Antigen , Curcumin , Immune Checkpoint Inhibitors , Immunotherapy , Urinary Bladder Neoplasms , Animals , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/therapy , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Curcumin/therapeutic use , Curcumin/administration & dosage , Mice , Immunotherapy/methods , Immune Checkpoint Inhibitors/therapeutic use , Humans , Cell Line, Tumor , Female , Colitis/chemically induced , Colitis/immunology , Colitis/drug therapy , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Drug Delivery Systems , Disease Models, Animal , Mice, Inbred C57BL , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/therapeutic use , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology
14.
Int Immunopharmacol ; 137: 112512, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38897123

ABSTRACT

OBJECTIVE: This study aims to disclose how loss of fucosyltransferase 2 (Fut2) impacts intestinal inflammation through cGAS-STING pathway that is closely associated with gut microbiota, and which microbial metabolite improves colitis in Fut2 deficiency. METHODS: Chronic colitis was induced in intestinal epithelial Fut2 knock out mice (Fut2△IEC), whose intestinal inflammation and activity of cGAS-STING pathway were evaluated. 16S rRNA sequencing and metabolomics were performed using intestinal samples. 2-oxindole was used to treat RAW264.7 cells and Fut2△IEC mice with colitis (Fut2△IEC-DSS) to investigate the effect of 2-oxindole on cGAS-STING response and intestinal inflammation. RESULTS: Fut2 loss exacerbated chronic colitis in mice, manifested by declined body weight, reduced colon length, increased disease activity index (DAI) and more colon injury in Fut2△IEC-DSS mice compared with WT-DSS (wild type mice with colitis). Lack of Fut2 promoted activation of cGAS-STING pathway. Fut2 deficiency had a primary impact on colonic microbiota, as shown by alteration of microbial diversity and structure, as well as decreased Lactobacillus. Metabolic structure and tryptophan metabolism in colonic luminal microbiota were also influenced by Fut2 loss. Fut2 deficiency also led to decreased levels of aryl hydrocarbon receptor (AHR) and its ligand 2-oxindole derived from tryptophan metabolism. 2-oxindole compromised cGAS-STING response through activating AHR in macrophages, and protected against intestinal inflammation and overactive cGAS-STING pathway in Fut2△IEC-DSS mice. CONCLUSION: Fut2 deficiency promotes cGAS-STING pathway through suppressing 2-oxindole-AHR axis, ultimately facilitating the susceptibility to chronic colitis.


Subject(s)
Colitis , Fucosyltransferases , Gastrointestinal Microbiome , Membrane Proteins , Mice, Knockout , Nucleotidyltransferases , Oxindoles , Signal Transduction , Animals , Mice , Colitis/chemically induced , Colitis/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Fucosyltransferases/deficiency , RAW 264.7 Cells , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Mice, Inbred C57BL , Male , Colon/pathology , Colon/immunology , Colon/metabolism , Chronic Disease , Disease Models, Animal , Humans , Dextran Sulfate
15.
NPJ Syst Biol Appl ; 10(1): 69, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38914538

ABSTRACT

Single-cell-based methods such as flow cytometry or single-cell mRNA sequencing (scRNA-seq) allow deep molecular and cellular profiling of immunological processes. Despite their high throughput, however, these measurements represent only a snapshot in time. Here, we explore how longitudinal single-cell-based datasets can be used for deterministic ordinary differential equation (ODE)-based modelling to mechanistically describe immune dynamics. We derived longitudinal changes in cell numbers of colonic cell types during inflammatory bowel disease (IBD) from flow cytometry and scRNA-seq data of murine colitis using ODE-based models. Our mathematical model generalised well across different protocols and experimental techniques, and we hypothesised that the estimated model parameters reflect biological processes. We validated this prediction of cellular turnover rates with KI-67 staining and with gene expression information from the scRNA-seq data not used for model fitting. Finally, we tested the translational relevance of the mathematical model by deconvolution of longitudinal bulk mRNA-sequencing data from a cohort of human IBD patients treated with olamkicept. We found that neutrophil depletion may contribute to IBD patients entering remission. The predictive power of IBD deterministic modelling highlights its potential to advance our understanding of immune dynamics in health and disease.


Subject(s)
Inflammatory Bowel Diseases , Single-Cell Analysis , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Single-Cell Analysis/methods , Humans , Mice , Animals , Flow Cytometry/methods , Colitis/genetics , Colitis/immunology , Longitudinal Studies
16.
Nat Immunol ; 25(8): 1383-1394, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38942990

ABSTRACT

The immunological mechanisms underlying chronic colitis are poorly understood. T follicular helper (TFH) cells are critical in helping B cells during germinal center reactions. In a T cell transfer colitis model, a lymphoid structure composed of mature dendritic cells (DCs) and TFH cells was found within T cell zones of colonic lymphoid follicles. TFH cells were required for mature DC accumulation, the formation of DC-T cell clusters and colitis development. Moreover, DCs promoted TFH cell differentiation, contributing to colitis development. A lineage-tracing analysis showed that, following migration to the lamina propria, TFH cells transdifferentiated into long-lived pathogenic TH1 cells, promoting colitis development. Our findings have therefore demonstrated the reciprocal regulation of TFH cells and DCs in colonic lymphoid follicles, which is critical in chronic colitis pathogenesis.


Subject(s)
Cell Differentiation , Colitis , Dendritic Cells , T Follicular Helper Cells , Animals , Dendritic Cells/immunology , Colitis/immunology , Colitis/pathology , T Follicular Helper Cells/immunology , Mice , Cell Differentiation/immunology , Mice, Inbred C57BL , Disease Models, Animal , Th1 Cells/immunology , Colon/immunology , Colon/pathology , Mice, Knockout , Germinal Center/immunology , Mice, Transgenic
17.
Cell Mol Immunol ; 21(8): 842-855, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38918576

ABSTRACT

The expression of clock genes has been observed to be impaired in biopsies from patients with inflammatory bowel disease (IBD). Disruption of circadian rhythms, which occurs in shift workers, has been linked to an increased risk of gastrointestinal diseases, including IBD. The peripheral circadian clock in intestinal epithelial cells (IECs) was previously shown to balance gastrointestinal homeostasis by regulating the microbiome. Here, we demonstrated that the intestinal clock is disrupted in an IBD-relevant mouse model (IL-10-/-). A lack of the intestinal clock gene (Bmal1) in intestinal epithelial cells (IECs) in a chemically and a novel genetically induced colitis model (DSS, Bmal1IEC-/-xIL-10-/-) promoted colitis and dramatically reduced survival rates. Germ-free Bmal1IEC-/- mice colonized with disease-associated microbiota from IL-10-/- mice exhibited increased inflammatory responses, highlighting the importance of the local intestinal clock for microbiota-induced IBD development. Targeting the intestinal clock directly by timed restricted feeding (RF) in IL-10-/- mice restored intestinal clock functions, including immune cell recruitment and microbial rhythmicity; improved inflammatory responses; dramatically enhanced survival rates and rescued the histopathological phenotype. In contrast, RF failed to improve IBD symptoms in Bmal1IEC-/-xIL-10-/- mice, demonstrating the significance of the intestinal clock in determining the beneficial effect of RF. Overall, we provide evidence that intestinal clock dysfunction triggers host immune imbalance and promotes the development and progression of IBD-like colitis. Enhancing intestinal clock function by RF modulates the pathogenesis of IBD and thus could become a novel strategy to ameliorate symptoms in IBD patients.


Subject(s)
ARNTL Transcription Factors , Circadian Clocks , Colitis , Interleukin-10 , Mice, Knockout , Animals , Circadian Clocks/genetics , Colitis/pathology , Colitis/immunology , Interleukin-10/metabolism , Interleukin-10/genetics , Mice , ARNTL Transcription Factors/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/deficiency , Mice, Inbred C57BL , Inflammation/pathology , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/immunology , Disease Models, Animal , Intestinal Mucosa/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Gastrointestinal Microbiome , Circadian Rhythm , Intestines/pathology , Intestines/microbiology , Intestines/immunology
18.
J Cancer Res Clin Oncol ; 150(5): 243, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38717677

ABSTRACT

Colitis-associated colorectal cancer has been a hot topic in public health issues worldwide. Numerous studies have demonstrated the significance of myeloid-derived suppressor cells (MDSCs) in the progression of this ailment, but the specific mechanism of their role in the transformation of inflammation to cancer is unclear, and potential therapies targeting MDSC are also unclear. This paper outlines the possible involvement of MDSC to the development of colitis-associated colorectal cancer. It also explores the immune and other relevant roles played by MDSC, and collates relevant targeted therapies against MDSC. In addition, current targeted therapies for colorectal cancer are analyzed and summarized.


Subject(s)
Colitis-Associated Neoplasms , Colorectal Neoplasms , Myeloid-Derived Suppressor Cells , Humans , Myeloid-Derived Suppressor Cells/immunology , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Animals , Colitis/complications , Colitis/immunology
19.
Commun Biol ; 7(1): 527, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714733

ABSTRACT

Macrophages are versatile cells of the innate immune system that work by altering their pro- or anti-inflammatory features. Their dysregulation leads to inflammatory disorders such as inflammatory bowel disease. We show that macrophage-specific upregulation of the clock output gene and transcription factor E4BP4 reduces the severity of colitis in mice. RNA-sequencing and single-cell analyses of macrophages revealed that increased expression of E4BP4 leads to an overall increase in expression of anti-inflammatory genes including Il4ra with a concomitant reduction in pro-inflammatory gene expression. In contrast, knockout of E4BP4 in macrophages leads to increased proinflammatory gene expression and decreased expression of anti-inflammatory genes. ChIP-seq and ATAC-seq analyses further identified Il4ra as a target of E4BP4, which drives anti-inflammatory polarization in macrophages. Together, these results reveal a critical role for E4BP4 in regulating macrophage inflammatory phenotypes and resolving inflammatory bowel diseases.


Subject(s)
Colitis , Macrophages , Animals , Macrophages/immunology , Macrophages/metabolism , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colitis/chemically induced , Mice , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Mice, Knockout , Phenotype , Mice, Inbred C57BL , Disease Models, Animal , Severity of Illness Index , Male , Inflammation/genetics , Inflammation/metabolism
20.
Int Immunopharmacol ; 136: 112325, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38820960

ABSTRACT

BACKGROUND: Although the pathogenesis of inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), has not been fully elucidated, accumulating researches suggest that intestinal microbiota imbalance contributes to the development of IBD in patients and animal models. RDP58, a peptide-based computer-assisted rational design, has been demonstrated to be effective in protecting against a wide range of autoimmune and inflammatory diseases. However, the underlying mechanism by which RDP58 protects against IBD mediated by intestinal microbiota has yet to be elucidated. METHODS: The colitis model was induced by continuously administering 2.5 % (wt/vol) dextran sodium sulfate (DSS) solution for 7 days. The manifestations of colon inflammation were assessed via daily weight changes, colon length, tumor necrosis factor-alpha (TNF-α) level, disease activity index (DAI) score, pathology score, and intestinal barrier permeability. Intestinal microbiota analysis was carried out by 16S-rRNA sequencing. Colonic short chain fatty acids (SCFAs) and regulatory T cells (Tregs) were also detected. To further confirm the protective effect of RDP58 on intestinal microbiota, broad-spectrum antibiotic cocktail (ABX) treatment and fecal microbial transplantation (FMT) experiment were performed. RESULTS: Oral administration of RDP58 ameliorated DSS-induced mice colitis by altering the diversity and composition of intestinal microbiota. Notably, RDP58 significantly upregulated SCFAs-producing microbiota, thereby promoting the generation of Tregs. ABX and FMT were performed to verify the above mechanism. CONCLUSIONS: RDP58 ameliorated DSS-induced colitis through altering intestinal microbiota and enhancing SCFAs and Tregs production in intestinal microbiota dependent manner, potentially provide a novel therapy for IBD.


Subject(s)
Colitis , Dextran Sulfate , Gastrointestinal Microbiome , Mice, Inbred C57BL , T-Lymphocytes, Regulatory , Animals , Gastrointestinal Microbiome/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/microbiology , Colitis/immunology , Administration, Oral , T-Lymphocytes, Regulatory/immunology , Mice , Disease Models, Animal , Colon/pathology , Colon/microbiology , Colon/drug effects , Colon/immunology , Male , Tumor Necrosis Factor-alpha/metabolism , Fecal Microbiota Transplantation , Humans , Fatty Acids, Volatile/metabolism , Oligopeptides
SELECTION OF CITATIONS
SEARCH DETAIL