Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.402
Filter
1.
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
2.
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
3.
Cancer Cell ; 42(5): 797-814.e15, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38744246

ABSTRACT

The success of checkpoint inhibitors (CPIs) for cancer has been tempered by immune-related adverse effects including colitis. CPI-induced colitis is hallmarked by expansion of resident mucosal IFNγ cytotoxic CD8+ T cells, but how these arise is unclear. Here, we track CPI-bound T cells in intestinal tissue using multimodal single-cell and subcellular spatial transcriptomics (ST). Target occupancy was increased in inflamed tissue, with drug-bound T cells located in distinct microdomains distinguished by specific intercellular signaling and transcriptional gradients. CPI-bound cells were largely CD4+ T cells, including enrichment in CPI-bound peripheral helper, follicular helper, and regulatory T cells. IFNγ CD8+ T cells emerged from both tissue-resident memory (TRM) and peripheral populations, displayed more restricted target occupancy profiles, and co-localized with damaged epithelial microdomains lacking effective regulatory cues. Our multimodal analysis identifies causal pathways and constitutes a resource to inform novel preventive strategies.


Subject(s)
Colitis , Immune Checkpoint Inhibitors , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/pharmacology , Humans , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Animals , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Interferon-gamma/metabolism , Female , Single-Cell Analysis , Mice
4.
Clin Transl Sci ; 17(5): e13821, 2024 May.
Article in English | MEDLINE | ID: mdl-38742709

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by a chronically dysregulated immune response in the gastrointestinal tract. Bone marrow multipotent mesenchymal stromal cells have an important immunomodulatory function and support regeneration of inflamed tissue by secretion of soluble factors as well as through direct local differentiation. CXCR4 is the receptor for CXCL12 (SDF-1, stromal-derived factor-1) and has been shown to be the main chemokine receptor, required for homing of MSCs. Increased expression of CXCL12 by inflamed intestinal tissue causes constitutive inflammation by attracting lymphocytes but can also be used to direct MSCs to sites of injury/inflammation. Trypsin is typically used to dissociate MSCs into single-cell suspensions but has also been shown to digest surface CXCR4. Here, we assessed the regenerative effects of CXCR4high and CXCR4low MSCs in an immune-deficient mouse model of DSS-induced colitis. We found that transplantation of MSCs resulted in clinical improvement and histological recovery of intestinal epithelium. In contrary to our expectations, the levels of CXCR4 on transplanted MSCs did not affect their regenerative supporting potential, indicating that paracrine effects of MSCs may be largely responsible for their regenerative/protective effects.


Subject(s)
Colitis , Disease Models, Animal , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mice, Inbred C57BL , Receptors, CXCR4 , Regeneration , Animals , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Mesenchymal Stem Cells/metabolism , Colitis/chemically induced , Colitis/pathology , Colitis/immunology , Colitis/therapy , Colitis/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mice , Dextran Sulfate , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/immunology , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Bone Marrow Cells/metabolism
6.
Nat Med ; 30(5): 1349-1362, 2024 May.
Article in English | MEDLINE | ID: mdl-38724705

ABSTRACT

Immune checkpoint inhibitor (ICI) therapy has revolutionized oncology, but treatments are limited by immune-related adverse events, including checkpoint inhibitor colitis (irColitis). Little is understood about the pathogenic mechanisms driving irColitis, which does not readily occur in model organisms, such as mice. To define molecular drivers of irColitis, we used single-cell multi-omics to profile approximately 300,000 cells from the colon mucosa and blood of 13 patients with cancer who developed irColitis (nine on anti-PD-1 or anti-CTLA-4 monotherapy and four on dual ICI therapy; most patients had skin or lung cancer), eight controls on ICI therapy and eight healthy controls. Patients with irColitis showed expanded mucosal Tregs, ITGAEHi CD8 tissue-resident memory T cells expressing CXCL13 and Th17 gene programs and recirculating ITGB2Hi CD8 T cells. Cytotoxic GNLYHi CD4 T cells, recirculating ITGB2Hi CD8 T cells and endothelial cells expressing hypoxia gene programs were further expanded in colitis associated with anti-PD-1/CTLA-4 therapy compared to anti-PD-1 therapy. Luminal epithelial cells in patients with irColitis expressed PCSK9, PD-L1 and interferon-induced signatures associated with apoptosis, increased cell turnover and malabsorption. Together, these data suggest roles for circulating T cells and epithelial-immune crosstalk critical to PD-1/CTLA-4-dependent tolerance and barrier function and identify potential therapeutic targets for irColitis.


Subject(s)
Colitis , Immune Checkpoint Inhibitors , Intestinal Mucosa , Single-Cell Analysis , Humans , Immune Checkpoint Inhibitors/adverse effects , Colitis/chemically induced , Colitis/immunology , Colitis/genetics , Colitis/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Female , Male , Gene Expression Profiling , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Aged , Transcriptome , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Colon/pathology , Colon/immunology , Colon/drug effects , Epithelial Cells/immunology , Epithelial Cells/drug effects , Epithelial Cells/pathology
7.
Int Immunopharmacol ; 134: 112234, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38739976

ABSTRACT

Ulcerative colitis, a chronic inflammatory condition affecting the rectum and colon to varying degrees, is linked to a dysregulated immune response and the microbiota. Sodium (aS,9R)-3-hydroxy-16,17-dimethoxy-15-oxidotricyclo[12.3.1.12,6]nonadeca-1(18),2,4,6(19),14,16-hexene-9-yl sulfate hydrate (SDH) emerges as a novel diarylheptane compound aimed at treating inflammatory bowel diseases. However, the mechanisms by which SDH modulates these conditions remain largely unknown. In this study, we assessed SDH's impact on the clinical progression of dextran sodium sulfate (DSS)-induced ulcerative colitis. Our results demonstrated that SDH significantly mitigated the symptoms of DSS-induced colitis, reflected in reduced disease activity index scores, alleviation of weight loss, shortening of the colorectum, and reduction in spleen swelling. Notably, SDH decreased the proportion of Th1/Th2/Th17 cells and normalized inflammatory cytokine levels in the colon. Furthermore, SDH treatment modified the gut microbial composition in mice with colitis, notably decreasing Bacteroidetes and Proteobacteria populations while substantially increasing Firmicutes, Actinobacteria, and Patescibacteria. In conclusion, our findings suggest that SDH may protect the colon from DSS-induced colitis through the regulation of Th1/Th2/Th17 cells and gut microbiota, offering novel insights into SDH's therapeutic potential.


Subject(s)
Colitis, Ulcerative , Dextran Sulfate , Diarylheptanoids , Gastrointestinal Microbiome , Mice, Inbred C57BL , Animals , Gastrointestinal Microbiome/drug effects , Mice , Diarylheptanoids/pharmacology , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Colitis, Ulcerative/microbiology , Colon/drug effects , Colon/immunology , Colon/pathology , Colon/microbiology , Cytokines/metabolism , Disease Models, Animal , Colitis/chemically induced , Colitis/drug therapy , Colitis/immunology , Colitis/microbiology , Male , Th1 Cells/immunology , Th1 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/drug effects , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Th2 Cells/immunology , Th2 Cells/drug effects , Humans
8.
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
9.
Int Immunopharmacol ; 135: 112298, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38776854

ABSTRACT

BACKGROUND: Excessive activation of colonic fibroblasts and differentiation of T helper 17 (Th17) cells are the key steps for intestinal fibrogenesis in the process of inflammatory bowel disease (IBD). Although both transforming growth factor-beta (TGF-ß)/Mothers Against Decapentaplegic Homolog (SMAD) 3-induced fibroblasts activation and interleukin (IL)-6/signal transducer and activator of transcription (STAT) 3-induced Th17 differentiation have been well studied, the crosstalk between fibroblasts and Th17 cells in the process of intestinal fibrogenesis needs to be unveiled. METHODS: In this study, the activation of colonic fibroblasts was induced with dextran sulfate sodium salt (DSS) and TGF-ß in vivo and in vitro respectively. P-SMAD3 and its downstream targets were quantified using RT-PCR, western blot and immunofluorescence. The differentiation of programmed death 1 (PD-1) + Th17 and activation of fibroblasts were quantified by FACS. PD-1+ Th17 cells and fibroblasts were co-cultured and cytokines in the supernatant were tested by ELISA. The anti-fibrosis effects of different chemical compounds were validated in vitro and further confirmed in vivo. RESULTS: The colonic fibroblasts were successfully activated by DSS and TGF-ß in vivo and in vitro respectively, as activation markers of fibroblasts (p-SMAD3 and its downstream targets such as Acta2, Col1a1 and Ctgf) were significantly increased. The activated fibroblasts produced more IL-6 compared with their inactivated counterparts in vivo and in vitro. The proinflammatory cytokine IL-6 induced PD-1+ Th17 differentiation and TGF-ß that in return promoted the activation of colonic fibroblasts. Fraxinellone inhibited TGF-ß+ PD-1+ Th17 cells via deactivating STAT3. CONCLUSIONS: The reciprocal stimulation constructed a circuit of PD-1+ Th17 cells and fibroblasts that accelerated the fibrosis process. Fraxinellone was selected as the potential inhibitor of the circuit of PD-1+ Th17 cells and fibroblasts in vivo and in vitro. Inhibiting the circuit of PD-1+ Th17 cells and fibroblasts could be a promising strategy to alleviate intestinal fibrosis.


Subject(s)
Colitis , Dextran Sulfate , Fibroblasts , Fibrosis , Mice, Inbred C57BL , Th17 Cells , Animals , Fibroblasts/drug effects , Th17 Cells/immunology , Th17 Cells/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/immunology , Mice , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Colon/pathology , Colon/drug effects , Colon/immunology , Cells, Cultured , Smad3 Protein/metabolism , Male , Transforming Growth Factor beta/metabolism , Humans , Signal Transduction/drug effects , Cell Differentiation/drug effects
10.
Microbiome ; 12(1): 76, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38649950

ABSTRACT

BACKGROUND: The etiology of inflammatory bowel disease (IBD) is unclear but involves both genetics and environmental factors, including the gut microbiota. Indeed, exacerbated activation of the gastrointestinal immune system toward the gut microbiota occurs in genetically susceptible hosts and under the influence of the environment. For instance, a majority of IBD susceptibility loci lie within genes involved in immune responses, such as caspase recruitment domain member 9 (Card9). However, the relative impacts of genotype versus microbiota on colitis susceptibility in the context of CARD9 deficiency remain unknown. RESULTS: Card9 gene directly contributes to recovery from dextran sodium sulfate (DSS)-induced colitis by inducing the colonic expression of the cytokine IL-22 and the antimicrobial peptides Reg3ß and Reg3γ independently of the microbiota. On the other hand, Card9 is required for regulating the microbiota capacity to produce AhR ligands, which leads to the production of IL-22 in the colon, promoting recovery after colitis. In addition, cross-fostering experiments showed that 5 weeks after weaning, the microbiota transmitted from the nursing mother before weaning had a stronger impact on the tryptophan metabolism of the pups than the pups' own genotype. CONCLUSIONS: These results show the role of CARD9 and its effector IL-22 in mediating recovery from DSS-induced colitis in both microbiota-independent and microbiota-dependent manners. Card9 genotype modulates the microbiota metabolic capacity to produce AhR ligands, but this effect can be overridden by the implantation of a WT or "healthy" microbiota before weaning. It highlights the importance of the weaning reaction occurring between the immune system and microbiota for host metabolism and immune functions throughout life. A better understanding of the impact of genetics on microbiota metabolism is key to developing efficient therapeutic strategies for patients suffering from complex inflammatory disorders. Video Abstract.


Subject(s)
CARD Signaling Adaptor Proteins , Colitis , Dextran Sulfate , Gastrointestinal Microbiome , Interleukin-22 , Interleukins , Pancreatitis-Associated Proteins , Animals , CARD Signaling Adaptor Proteins/genetics , Colitis/microbiology , Colitis/genetics , Colitis/immunology , Mice , Pancreatitis-Associated Proteins/genetics , Interleukins/genetics , Interleukins/metabolism , Mice, Knockout , Genetic Predisposition to Disease , Disease Models, Animal , Mice, Inbred C57BL , Colon/microbiology , Colon/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Female , Male
11.
Discov Med ; 36(183): 778-787, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38665026

ABSTRACT

BACKGROUND: Tropomyosin 2 (TPM2) has been linked to the advancement of various tumor types, exhibiting distinct impacts on tumor progression. In our investigation, the primary objective was to identify the potential involvement of TPM2 in the development of colitis-associated cancer (CAC) using a mice model. METHODS: This study used lentiviral vector complex for TPM2 knockdown (sh-TPM2) and the corresponding negative control lentiviral vector complex (sh-NC) for genetic interference in mice. CAC was induced in mice using azoxymethane (AOM) and dextran sulfate sodium salt (DSS). This study included 6 groups of mice models: Control, Control+sh-NC, Control+sh-TPM2, CAC, CAC+sh-NC, and CAC+sh-TPM2. Subsequently, colon tissues were collected and assessed using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) for TPM2 mRNA levels and flow cytometry for infiltrating immune cells. Tumor number, size, and weight within colon tissues from CAC mice were measured and recorded. The hematoxylin-eosin staining was used for observing tissue pathology changes. The intestinal epithelial cells (IECs) were isolated and analyzed for cell proliferation. This analysis included examining the levels of 5-bromo-2-deoxyuridine (BrdU) and Ki-67 using immunohistochemistry. Additionally, the mRNA levels of proliferating cell nuclear antigen (PCNA) and Ki-67 were detected by qRT-PCR. This study also investigated the activation of the c-Jun N-terminal kinase (JNK) pathway using western blot analysis. Immunogenicity analyses were conducted using immunohistochemistry for F4/80 and flow cytometry. RESULTS: In 8-week-old mice, AOM injections and three cycles of DSS treatment induced TPM2 upregulation in tumor tissues compared to normal tissues (p < 0.05). Fluorescence-activated cell sorting (FACS)-isolated lamina CAC adenomas revealed macrophages and dendritic cells as primary TPM2 contributors (p < 0.001). Lentiviral TPM2 gene knockdown significantly reduced tumor numbers and sizes in CAC mice (p < 0.01, and p < 0.001), without invasive cancer cells. TPM2 suppression resulted in decreased IEC proliferation (p < 0.001) and reduced PCNA and Ki-67 expression (p < 0.05). Western blot analysis indicated reduced JNK pathway activation in TPM2-knockdown CAC mice (p < 0.05, p < 0.001). TPM2 knockdown decreased tumor-associated macrophage infiltration (p < 0.01) and increased CD3+ and CD8+ T cells (p < 0.01, and p < 0.001), with increased levels of regulator of inflammatory cytokines (CD44+, CD107a+) (p < 0.01, and p < 0.001), decreased levels of PD-1+ and anti-inflammatory factor (IL10+) (p < 0.01, and p < 0.001). CONCLUSIONS: Our results demonstrated that TPM2 knockdown suppressed the proliferation of CAC IECs, enhanced immune suppression on CAC IECs, and inhibited the JNK signaling pathway within the framework of CAC. These findings suggest TPM2 can serve as a potential therapeutic target for CAC treatment.


Subject(s)
Cell Proliferation , Colitis-Associated Neoplasms , MAP Kinase Signaling System , Tropomyosin , Animals , Humans , Male , Mice , Azoxymethane/toxicity , Colitis/chemically induced , Colitis/pathology , Colitis/complications , Colitis/immunology , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/immunology , Colitis-Associated Neoplasms/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , MAP Kinase Signaling System/immunology , Mice, Inbred C57BL , Tropomyosin/metabolism , Tropomyosin/immunology , Tropomyosin/genetics
12.
Proc Natl Acad Sci U S A ; 121(18): e2312111121, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38657041

ABSTRACT

Class II histone deacetylases (HDACs) are important in regulation of gene transcription during T cell development. However, our understanding of their cell-specific functions is limited. In this study, we reveal that class IIa Hdac4 and Hdac7 (Hdac4/7) are selectively induced in transcription, guiding the lineage-specific differentiation of mouse T-helper 17 (Th17) cells from naive CD4+ T cells. Importantly, Hdac4/7 are functionally dispensable in other Th subtypes. Mechanistically, Hdac4 interacts with the transcription factor (TF) JunB, facilitating the transcriptional activation of Th17 signature genes such as Il17a/f. Conversely, Hdac7 collaborates with the TF Aiolos and Smrt/Ncor1-Hdac3 corepressors to repress transcription of Th17 negative regulators, including Il2, in Th17 cell differentiation. Inhibiting Hdac4/7 through pharmacological or genetic methods effectively mitigates Th17 cell-mediated intestinal inflammation in a colitis mouse model. Our study uncovers molecular mechanisms where HDAC4 and HDAC7 function distinctively yet cooperatively in regulating ordered gene transcription during Th17 cell differentiation. These findings suggest a potential therapeutic strategy of targeting HDAC4/7 for treating Th17-related inflammatory diseases, such as ulcerative colitis.


Subject(s)
Cell Differentiation , Colitis , Histone Deacetylases , Nuclear Receptor Co-Repressor 1 , Th17 Cells , Animals , Th17 Cells/cytology , Th17 Cells/metabolism , Th17 Cells/immunology , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Mice , Colitis/genetics , Colitis/metabolism , Colitis/immunology , Transcription, Genetic , Transcription Factors/metabolism , Transcription Factors/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Nuclear Receptor Co-Repressor 2/genetics , Interleukin-17/metabolism , Gene Expression Regulation , Mice, Inbred C57BL , Humans , Repressor Proteins/metabolism , Repressor Proteins/genetics , Interleukin-2/metabolism
14.
Cells ; 13(8)2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38667290

ABSTRACT

Excessive secretion of pro-inflammatory cytokines leads to the disruption of intestinal barrier in inflammatory bowel disease (IBD). The inflammatory cytokine tumor necrosis factor alpha (TNFα) induces the assembly of the NLRP3 inflammasome, resulting in the augmented secretion of inflammatory cytokines implicated in the pathogenesis of inflammatory bowel disease (IBD). TNFα has also been known to induce the formation of immunoproteasome (IP), which incorporates immunosubunits LMP2, LMP7, and MECL-1. Inhibition of IP activity using the IP subunit LMP2-specific inhibitor YU102, a peptide epoxyketone, decreased the protein levels of NLRP3 and increased the K48-linked polyubiquitination levels of NLRP3 in TNFα-stimulated intestinal epithelial cells. We observed that inhibition of IP activity caused an increase in the protein level of the ubiquitin E3 ligase, tripartite motif-containing protein 31 (TRIM31). TRIM31 facilitated K48-linked polyubiquitination and proteasomal degradation of NLRP3 with an enhanced interaction between NLRP3 and TRIM31 in intestinal epithelial cells. In addition, IP inhibition using YU102 ameliorated the symptoms of colitis in the model mice inflicted with dextran sodium sulfate (DSS). Administration of YU102 in the DSS-treated colitis model mice caused suppression of the NLRP3 protein levels and accompanied inflammatory cytokine release in the intestinal epithelium. Taken together, we demonstrated that inhibiting IP under inflammatory conditions induces E3 ligase TRIM31-mediated NLRP3 degradation, leading to attenuation of the NLRP3 inflammatory response that triggers disruption of intestinal barrier.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Proteasome Endopeptidase Complex , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Ubiquitination , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Ubiquitin-Protein Ligases/metabolism , Tripartite Motif Proteins/metabolism , Inflammasomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Mice , Humans , Ubiquitination/drug effects , Mice, Inbred C57BL , Colitis/chemically induced , Colitis/pathology , Colitis/metabolism , Colitis/immunology , Tumor Necrosis Factor-alpha/metabolism , Dextran Sulfate , Disease Models, Animal
15.
Cell Mol Immunol ; 21(6): 589-603, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38649449

ABSTRACT

Inflammatory bowel disease (IBD) is prevalent, and no satisfactory therapeutic options are available because the mechanisms underlying its development are poorly understood. In this study, we discovered that increased expression of methyltransferase-like 3 (METTL3) in macrophages was correlated with the development of colitis and that depletion of METTL3 in macrophages protected mice against dextran sodium sulfate (DSS)-induced colitis. Mechanistic characterization indicated that METTL3 depletion increased the YTHDF3-mediated expression of phosphoglycolate phosphatase (PGP), which resulted in glucose metabolism reprogramming and the suppression of CD4+ T helper 1 (Th1) cell differentiation. Further analysis revealed that glucose metabolism contributed to the ability of METTL3 depletion to ameliorate colitis symptoms. In addition, we developed two potent small molecule METTL3 inhibitors, namely, F039-0002 and 7460-0250, that strongly ameliorated DSS-induced colitis. Overall, our study suggests that METTL3 plays crucial roles in the progression of colitis and highlights the potential of targeting METTL3 to attenuate intestinal inflammation for the treatment of colitis.


Subject(s)
Colitis , Dextran Sulfate , Macrophages , Methyltransferases , Mice, Inbred C57BL , Animals , Methyltransferases/metabolism , Colitis/chemically induced , Colitis/pathology , Colitis/immunology , Macrophages/metabolism , Macrophages/drug effects , Macrophages/immunology , Mice , Inflammation/pathology , Th1 Cells/immunology , Cell Differentiation/drug effects , Humans , Intestines/pathology , Disease Models, Animal
16.
Front Immunol ; 15: 1366489, 2024.
Article in English | MEDLINE | ID: mdl-38660314

ABSTRACT

Cancer ranks among the foremost causes of mortality worldwide, posing a significant threat to human lives. The advent of tumor immunotherapy has substantially transformed the therapeutic landscape for numerous advanced malignancies, notably non-small cell lung cancer and melanoma. However, as immune checkpoint inhibitors (ICIs) are increasingly applied in clinical settings, a spectrum of undesired reactions, termed immune-related adverse events (irAEs), has emerged. These adverse reactions are associated with immunotherapy and can result in varying degrees of harm to the human body. Among these reactions, Immune checkpoint inhibitor-induced colitis (ICIIC) stands out as one of the most prevalent clinical adverse events. In contemporary times, traditional Chinese medicine (TCM) has demonstrated remarkable efficacy in addressing various maladies. Consequently, investigating the potential application and mechanisms of Chinese medicine in countering immune checkpoint inhibitor-induced colitis assumes significant importance in the treatment of this condition.


Subject(s)
Colitis , Immune Checkpoint Inhibitors , Medicine, Chinese Traditional , Humans , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/therapeutic use , Colitis/chemically induced , Colitis/immunology , Colitis/therapy , Animals , Drugs, Chinese Herbal/adverse effects , Drugs, Chinese Herbal/therapeutic use , Immunotherapy/adverse effects , Immunotherapy/methods
17.
ACS Biomater Sci Eng ; 10(5): 3041-3056, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38623037

ABSTRACT

Oral immunization is a promising strategy for preventing and treating gastrointestinal (GI) infections and diseases, as it allows for direct access to the disease site. To elicit immune responses within the GI tract, however, there are many obstacles that oral vaccines must surmount, including proteolytic degradation and thick mucus barriers. Here, we employed a modular self-assembling peptide nanofiber platform to facilitate oral immunization against both peptide and small molecule epitopes. Synthesizing nanofibers with d-amino acids rendered them resistant to proteases in vitro, whereas l-amino acid nanofibers were rapidly degraded. Additionally, the inclusion of peptide sequences rich in proline, alanine, and serine (PAS), increased nanofiber muco-penetration, and accelerated nanofiber transport through the GI tract. Oral immunization with PASylated nanofibers and mucosal adjuvant generated local and systemic immune responses to a peptide epitope but only for l-amino acid nanofibers. Further, we were able to apply this design to also enable oral immunization against a small molecule epitope and illustrated the therapeutic and prophylactic effectiveness of these immunizations in mouse models of colitis. These findings demonstrate that supramolecular peptide self-assemblies have promise as oral vaccines and immunotherapies.


Subject(s)
Immunization , Nanofibers , Peptides , Animals , Administration, Oral , Nanofibers/chemistry , Peptides/immunology , Peptides/chemistry , Peptides/administration & dosage , Mice , Immunization/methods , Epitopes/immunology , Female , Mice, Inbred C57BL , Colitis/immunology , Colitis/prevention & control , Colitis/chemically induced
18.
Nat Immunol ; 25(5): 886-901, 2024 May.
Article in English | MEDLINE | ID: mdl-38609547

ABSTRACT

Intestinal immune responses to microbes are controlled by the cytokine IL-10 to avoid immune pathology. Here, we use single-cell RNA sequencing of colon lamina propria leukocytes (LPLs) along with RNA-seq and ATAC-seq of purified CD4+ T cells to show that the transcription factors Blimp-1 (encoded by Prdm1) and c-Maf co-dominantly regulate Il10 while negatively regulating proinflammatory cytokines in effector T cells. Double-deficient Prdm1fl/flMaffl/flCd4Cre mice infected with Helicobacter hepaticus developed severe colitis with an increase in TH1/NK/ILC1 effector genes in LPLs, while Prdm1fl/flCd4Cre and Maffl/flCd4Cre mice exhibited moderate pathology and a less-marked type 1 effector response. LPLs from infected Maffl/flCd4Cre mice had increased type 17 responses with increased Il17a and Il22 expression and an increase in granulocytes and myeloid cell numbers, resulting in increased T cell-myeloid-neutrophil interactions. Genes over-expressed in human inflammatory bowel disease showed differential expression in LPLs from infected mice in the absence of Prdm1 or Maf, revealing potential mechanisms of human disease.


Subject(s)
Colitis , Helicobacter hepaticus , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-maf , Animals , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Mice , Proto-Oncogene Proteins c-maf/genetics , Colitis/immunology , Colitis/genetics , Humans , Helicobacter hepaticus/immunology , Helicobacter Infections/immunology , Mice, Inbred C57BL , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/microbiology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/genetics , Gene Expression Regulation , Disease Models, Animal
19.
Cell Mol Immunol ; 21(6): 561-574, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38570588

ABSTRACT

Hyperactivation of the NLRP3 inflammasome has been implicated in the pathogenesis of numerous diseases. However, the precise molecular mechanisms that modulate the transcriptional regulation of NLRP3 remain largely unknown. In this study, we demonstrated that S-nitrosoglutathione reductase (GSNOR) deficiency in macrophages leads to significant increases in the Nlrp3 and Il-1ß expression levels and interleukin-1ß (IL-1ß) secretion in response to NLRP3 inflammasome stimulation. Furthermore, in vivo experiments utilizing Gsnor-/- mice revealed increased disease severity in both lipopolysaccharide (LPS)-induced septic shock and dextran sodium sulfate (DSS)-induced colitis models. Additionally, we showed that both LPS-induced septic shock and DSS-induced colitis were ameliorated in Gsnor-/- Nlrp3-/- double-knockout (DKO) mice. Mechanistically, GSNOR deficiency increases the S-nitrosation of mitogen-activated protein kinase 14 (MAPK14) at the Cys211 residue and augments MAPK14 kinase activity, thereby promoting Nlrp3 and Il-1ß transcription and stimulating NLRP3 inflammasome activity. Our findings suggested that GSNOR is a regulator of the NLRP3 inflammasome and that reducing the level of S-nitrosylated MAPK14 may constitute an effective strategy for alleviating diseases associated with NLRP3-mediated inflammation.


Subject(s)
Colitis , Dextran Sulfate , Inflammasomes , Interleukin-1beta , Lipopolysaccharides , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Mice , Aldehyde Oxidoreductases/metabolism , Aldehyde Oxidoreductases/genetics , Colitis/chemically induced , Colitis/pathology , Colitis/immunology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Macrophages/immunology , Nitrosation , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Shock, Septic/metabolism , Shock, Septic/chemically induced , Mitogen-Activated Protein Kinase 14/metabolism
20.
Am J Physiol Gastrointest Liver Physiol ; 326(5): G591-G606, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38469632

ABSTRACT

Ulcerative colitis (UC) is an idiopathic inflammatory disease of the large intestine, which impacts millions worldwide. Current interventions aimed at treating UC symptoms can have off-target effects, invoking the need for alternatives that may provide similar benefits with less unintended consequences. This study builds on our initial data, which showed that panaxynol-a novel, potent, bioavailable compound found in American ginseng-can suppress disease severity in murine colitis. Here we explore the underlying mechanisms by which panaxynol improves both chronic and acute murine colitis. Fourteen-week-old C57BL/6 female mice were either given three rounds of dextran sulfate sodium (DSS) in drinking water to induce chronic colitis or one round to induce acute colitis. Vehicle or panaxynol (2.5 mg/kg) was administered via oral gavage three times per week for the study duration. Consistent with our previous findings, panaxynol significantly (P < 0.05) improved the disease activity index and endoscopic scores in both models. Using the acute model to examine potential mechanisms, we show that panaxynol significantly (P < 0.05) reduced DSS-induced crypt distortion, goblet cell loss, and mucus loss in the colon. 16S Sequencing revealed panaxynol altered microbial composition to suppress colitis-enriched genera (i.e., Enterococcus, Eubacterium, and Ruminococcus). In addition, panaxynol significantly (P < 0.05) suppressed macrophages and induced regulatory T-cells in the colonic lamina propria. The beneficial effects of panaxynol on mucosal and crypt architecture, combined with its microbial and immune-mediated effects, provide insight into the mechanisms by which panaxynol suppresses murine colitis. Overall, this data is promising for the use of panaxynol to improve colitis in the clinic.NEW & NOTEWORTHY In the current study, we report that panaxynol ameliorates chemically induced murine colitis by improving colonic crypt and mucosal architecture, suppressing colitis-enriched microbes, reducing macrophages, and promoting the differentiation of regulatory T-cells in the colonic lamina propria. This study suggests that this novel natural compound may serve as a safe and effective treatment option for colitis patients.


Subject(s)
Colitis , Dextran Sulfate , Gastrointestinal Microbiome , Intestinal Mucosa , Mice, Inbred C57BL , Animals , Female , Mice , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/microbiology , Intestinal Mucosa/immunology , Gastrointestinal Microbiome/drug effects , Colitis/drug therapy , Colitis/chemically induced , Colitis/pathology , Colitis/immunology , Colitis/microbiology , Fatty Alcohols/pharmacology , Diynes/pharmacology , Disease Models, Animal , Colon/drug effects , Colon/pathology , Colon/immunology , Colon/microbiology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Colitis, Ulcerative/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...