Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.313
Filter
1.
Med Oncol ; 41(10): 240, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39231878

ABSTRACT

Interleukin-22, discovered in the year of 2000, is a pleiotropic Th17 cytokine from the IL-10 family of cytokines. IL-22 signals through the type 2 cytokine receptor complex IL-22R and predominantly activates STAT3. This pathway leads to the transcription of several different types of genes, giving IL-22 context-specific functions ranging from inducing antimicrobial peptide expression to target cell proliferation. In recent years, it has been shown that IL-22 is involved in the pathogenesis of neoplasia in some cancers through its pro-proliferative and anti-apoptotic effects. This review highlights studies with recent discoveries and conclusions drawn on IL-22 and its involvement and function in various cancers. Such a study may be helpful to better understand the role of IL-22 in cancer so that new treatment could be developed targeting IL-22.


Subject(s)
Interleukin-22 , Interleukins , Neoplasms , Humans , Interleukins/metabolism , Neoplasms/metabolism , Neoplasms/immunology , Neoplasms/pathology , Animals , Signal Transduction , STAT3 Transcription Factor/metabolism , Receptors, Interleukin/metabolism , Receptors, Interleukin/genetics
2.
Appl Microbiol Biotechnol ; 108(1): 469, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39298023

ABSTRACT

Prior research has indicated that the gut-lung-axis can be influenced by the intestinal microbiota, thereby impacting lung immunity. Rifaximin is a broad-spectrum antibacterial drug that can maintain the homeostasis of intestinal microflora. In this study, we established an influenza A virus (IAV)-infected mice model with or without rifaximin supplementation to investigate whether rifaximin could ameliorate lung injury induced by IAV and explore the molecular mechanism involved. Our results showed that IAV caused significant weight loss and disrupted the structure of the lung and intestine. The analysis results of 16S rRNA and metabolomics indicated a notable reduction in the levels of probiotics Lachnoclostridium, Ruminococcaceae_UCG-013, and tryptophan metabolites in the fecal samples of mice infected with IAV. In contrast, supplementation with 50 mg/kg rifaximin reversed these changes, including promoting the repair of the lung barrier and increasing the abundance of Muribaculum, Papillibacter and tryptophan-related metabolites content in the feces. Additionally, rifaximin treatment increased ILC3 cell numbers, IL-22 level, and the expression of RORγ and STAT-3 protein in the lung. Furthermore, our findings demonstrated that the administration of rifaximin can mitigate damage to the intestinal barrier while enhancing the expression of AHR, IDO-1, and tight junction proteins in the small intestine. Overall, our results provided that rifaximin alleviated the imbalance in gut microbiota homeostasis induced by IAV infection and promoted the production of tryptophan-related metabolites. Tryptophan functions as a signal to facilitate the activation and movement of ILC3 cells from the intestine to the lung through the AHR/STAT3/IL-22 pathway, thereby aiding in the restoration of the barrier. KEY POINTS: • Rifaximin ameliorated IAV infection-caused lung barrier injury and induced ILC3 cell activation. • Rifaximin alleviated IAV-induced gut dysbiosis and recovered tryptophan metabolism. • Tryptophan mediates rifaximin-induced ILC3 cell activation via the AHR/STAT3/IL-22 pathway.


Subject(s)
Gastrointestinal Microbiome , Influenza A virus , Lung , Orthomyxoviridae Infections , Rifaximin , Animals , Gastrointestinal Microbiome/drug effects , Rifaximin/therapeutic use , Mice , Lung/microbiology , Lung/drug effects , Orthomyxoviridae Infections/drug therapy , Influenza A virus/drug effects , Disease Models, Animal , RNA, Ribosomal, 16S/genetics , Interleukins/metabolism , Interleukins/genetics , Interleukin-22 , Mice, Inbred C57BL , Anti-Bacterial Agents/pharmacology , STAT3 Transcription Factor/metabolism , Feces/microbiology , Tryptophan/metabolism , Lung Injury/drug therapy , Probiotics/administration & dosage , Probiotics/pharmacology
3.
Cell Death Dis ; 15(9): 687, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300068

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) are essential for both pathogen defense and tissue homeostasis in the intestine. Dysfunction of ILC3s could lead to increased susceptibility to intestinal inflammation. However, the precise mechanisms governing the maintenance of intestinal ILC3s are yet to be fully elucidated. Here, we demonstrated that ferroptosis is vital for regulating the survival of intestinal ILC3. Ferroptosis-related genes, including GPX4, a key regulator of ferroptosis, were found to be upregulated in intestinal mucosal ILC3s from ulcerative colitis patients. Deletion of GPX4 resulted in a decrease in NKp46+ILC3 cell numbers, impaired production of IL-22 and IL-17A, and exacerbated intestinal inflammation in a T cell-independent manner. Our mechanistic studies revealed that GPX4-mediated ferroptosis in NKp46+ILC3 cells was regulated by the LCN2-p38-ATF4-xCT signaling pathway. Mice lacking LCN2 in ILC3s or administration of a p38 pathway inhibitor exhibited similar phenotypes of ILC3 and colitis to those observed in GPX4 conditional knock-out mice. These observations provide novel insights into therapeutic strategies for intestinal inflammation by modulating ILC3 ferroptosis.


Subject(s)
Ferroptosis , Inflammation , Natural Cytotoxicity Triggering Receptor 1 , Phospholipid Hydroperoxide Glutathione Peroxidase , Ferroptosis/genetics , Animals , Mice , Natural Cytotoxicity Triggering Receptor 1/metabolism , Natural Cytotoxicity Triggering Receptor 1/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Humans , Inflammation/pathology , Inflammation/metabolism , Lymphocytes/metabolism , Lymphocytes/immunology , Mice, Inbred C57BL , Mice, Knockout , Interleukin-22 , Immunity, Innate , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Signal Transduction , Intestines/pathology , Colitis, Ulcerative/pathology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/genetics , Antigens, Ly/metabolism
4.
Exp Dermatol ; 33(9): e15177, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39304339

ABSTRACT

Efficacy and safety of ritlecitinib (an oral JAK3/TEC family kinase inhibitor) were evaluated in patients with nonsegmental vitiligo (NSV) across Fitzpatrick skin types (FSTs). Patients with FST I-III ('light skin'; n = 247) and FST IV-VI ('dark skin'; n = 117) received once-daily ritlecitinib 50 mg (with/without 4-week loading dose), low-dose ritlecitinib or placebo for 24 weeks. At baseline, patients with light skin displayed higher CLM-1 and NCR1 serum levels than patients with dark skin (p < 0.05). At 24 weeks, ritlecitinib 50 mg improved the extent of depigmentation measured by percent change from baseline in facial-vitiligo area scoring index (placebo-adjusted mean difference [90% CI]) in patients with light (-15.2 [-24.7, -5.8]; p = 0.004) and dark (-37.4 [-50.3, -24.4]; p < 0.0001) skin, with continuous re-pigmentation through week 48. Treatment-emergent adverse events were similar across FSTs. At weeks 4 and 24, ritlecitinib 50 mg reduced CXCL11 serum levels (p < 0.001) in patients with light skin, whereas patients with dark skin had increased levels at week 4 (p = 0.05) and no significant change at week 24. Ritlecitinib 50 mg decreased IL-9 and IL-22 expression levels in dark skin compared with light skin (qPCR; p < 0.05). These differences in immune dysregulations may explain why NSV patients with dark skin respond to therapy earlier than patients with light skin.


Subject(s)
Biomarkers , Vitiligo , Humans , Vitiligo/drug therapy , Vitiligo/metabolism , Biomarkers/blood , Biomarkers/metabolism , Male , Female , Middle Aged , Skin Pigmentation/drug effects , Adult , Interleukins/metabolism , Interleukins/blood , Treatment Outcome , Double-Blind Method , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Interleukin-22
5.
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
6.
Mycopathologia ; 189(5): 85, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39283337

ABSTRACT

Malassezia, the most abundant fungal commensal on the mammalian skin, has been linked to several inflammatory skin diseases such as atopic dermatitis, seborrheic dermatitis and psoriasis. This study reveals that epicutaneous application with Malassezia globosa (M. globosa) triggers skin inflammation in mice. RNA-sequencing of the resulting mouse lesions indicates activation of Interleukin-17 (IL-17) signaling and T helper 17 (Th17) cells differentiation pathways by M. globosa. Furthermore, our findings demonstrate a significant upregulation of IL-23, IL-23R, IL-17A, and IL-22 expressions, along with an increase in the proportion of Th17 and pathogenic Th17 cells in mouse skin exposed to M. globosa. In vitro experiments illustrate that M. globosa prompts human primary keratinocytes to secrete IL-23 via TLR2/MyD88/NF-κB signaling. This IL-23 secretion by keratinocytes is shown to be adequate for inducing the differentiation of pathogenic Th17 cells in the skin. Overall, these results underscore the significant role of Malassezia in exacerbating skin inflammation by stimulating IL-23 secretion by keratinocytes and promoting the differentiation of pathogenic Th17 cells.


Subject(s)
Cell Differentiation , Interleukin-23 , Keratinocytes , Malassezia , Th17 Cells , Malassezia/immunology , Keratinocytes/microbiology , Keratinocytes/immunology , Keratinocytes/metabolism , Th17 Cells/immunology , Animals , Interleukin-23/metabolism , Humans , Mice , Signal Transduction , NF-kappa B/metabolism , Toll-Like Receptor 2/metabolism , Interleukin-17/metabolism , Skin/microbiology , Skin/pathology , Skin/immunology , Disease Models, Animal , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Cells, Cultured , Mice, Inbred C57BL , Interleukin-22
7.
Food Funct ; 15(18): 9563-9578, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39228354

ABSTRACT

This single blind placebo-controlled study has as its main objectives to investigate the influence of dark sweet cherries (DSC) consumption on obesity-related dysbiosis, metabolic endotoxemia, and intestinal permeability. Participants (>18 years old, BMI: 30-40 kg m-2) consumed 200 mL of DSC juice with 3 g of DSC powder (n = 19) or a placebo drink (n = 21) twice per day for 30 days. The gut microbiota abundance was investigated using 16S ribosomal RNA sequencing on fecal DNA. Metabolic endotoxemia was evaluated by measuring lipopolysaccharide-binding protein (LBP) in fasting plasma samples. Intestinal permeability was assessed using the lactulose/mannitol (L/M) test and by measuring regeneration islet-derived protein 4 (REG4), and interleukin-22 (IL-22) mRNA levels in stool samples. Results showed that DSC supplementation decreased the abundance of Anaerostipes hadrus (p = 0.02) and Blautia (p = 0.04), whose changes were significant in BMI ≥ 35 participants (p = 0.004 and p = 0.006, respectively). Additionally, DSC prevented the increase of Alistipes shahii (p = 0.005) and Bilophila (p = 0.01) compared to placebo. Notably, DSC intervention favored the abundance of bacteria supporting a healthy gut ecosystem such as Roseburia intestinalis (p = 0.01), Turicibacter (p = 0.01), and Bacteroides vulgatus (p = 0.003) throughout the intervention, along with Clostridium leptum (p = 0.03) compared to placebo. The LBP, L/M ratio, REG-4 and IL-22 mRNA levels remained unchanged in placebo and cherry groups, implying that participants did not experience alterations in intestinal permeability. These findings highlight the potential gut-health benefits of DSC and encourage future research among individuals with BMI ≥ 35 and increased intestinal permeability.


Subject(s)
Dietary Supplements , Endotoxemia , Feces , Gastrointestinal Microbiome , Obesity , Permeability , Prunus avium , Humans , Gastrointestinal Microbiome/drug effects , Male , Feces/microbiology , Obesity/microbiology , Obesity/metabolism , Obesity/diet therapy , Adult , Female , Middle Aged , Single-Blind Method , Interleukin-22 , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Intestinal Mucosa/metabolism , Young Adult , Intestinal Barrier Function
8.
Front Immunol ; 15: 1402834, 2024.
Article in English | MEDLINE | ID: mdl-39253083

ABSTRACT

Introduction: Group 3 innate lymphoid cells (ILC3s) are enriched in the intestinal mucosa and play important roles in host defense against infection and inflammatory diseases. Sirtuin 6 (SIRT6) is a nicotinamide adenine dinucleotide (NAD+)- dependent deacetylase and has been shown to control intestinal epithelial cell differentiation and survival. However, the role of SIRT6 in ILC3s remains unknown. Methods: To investigate the role of SIRT6 in gut ILC3s, we generated SIRT6 conditional knockout mice by crossing Rorccre and Sirt6flox/flox mice. Cell number and cytokine production was examined using flow cytometry. Citrobacter rodentium infection and dextran sodium sulfate-induced colitis models were used to determine the role of SIRT6 in gut defense. RT-qPCR, flow cytometry and immunohistochemistry were used to assess the intestinal inflammatory responses. Results: Here we show that SIRT6 inhibits IL-22 expression in intestinal ILC3s in a cell-intrinsic manner. Deletion of SIRT6 in ILC3s does not affect the cell numbers of total ILC3s and subsets, but results in increased IL-22 production. Furthermore, ablation of SIRT6 in ILC3s protects mice against Citrobacter rodentium infection and dextran sodium sulfate-induced colitis. Our results suggest that SIRT6 may play a role in ILC3 function by regulating gut immune responses against bacterial infection and inflammation. Discussion: Our finding provided insight into the relation of epigenetic regulators with IL-22 production and supplied a new perspective for a potential strategy against inflammatory bowel disease.


Subject(s)
Citrobacter rodentium , Colitis , Enterobacteriaceae Infections , Immunity, Innate , Interleukin-22 , Interleukins , Lymphocytes , Mice, Knockout , Sirtuins , Animals , Mice , Lymphocytes/immunology , Lymphocytes/metabolism , Interleukins/metabolism , Interleukins/immunology , Interleukins/genetics , Sirtuins/genetics , Sirtuins/metabolism , Colitis/immunology , Colitis/chemically induced , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Dextran Sulfate , Disease Models, Animal
9.
Cells ; 13(16)2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39195286

ABSTRACT

Atopic dermatitis (AD) is a prevalent and chronic inflammatory skin condition characterized by a multifaceted pathophysiology that gives rise to diverse clinical manifestations. The management of AD remains challenging due to the suboptimal efficacy of existing treatment options. Nonetheless, recent progress in elucidating the underlying mechanisms of the disease has facilitated the identification of new potential therapeutic targets and promising drug candidates. In this review, we summarize the newest data, considering multiple connections between IL-22 and AD. The presence of circulating IL-22 has been found to correlate with the severity of AD and is identified as a critical factor driving the inflammatory response associated with the condition. Elevated levels of IL-22 in patients with AD are correlated with increased proliferation of keratinocytes, alterations in the skin microbiota, and impaired epidermal barrier function. Collectively, these factors contribute to the manifestation of the characteristic symptoms observed in AD.


Subject(s)
Dermatitis, Atopic , Interleukin-22 , Interleukins , Dermatitis, Atopic/pathology , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , Humans , Interleukins/metabolism , Animals , Keratinocytes/metabolism , Skin/pathology , Skin/metabolism , Microbiota
10.
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
11.
Arch Dermatol Res ; 316(8): 561, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39177716

ABSTRACT

Circular RNAs (circRNAs) are demonstrated to be involved in psoriasis progression. CircRNAs can act as RNA-binding protein (RBP) sponges. Here, we investigated the action of circAKR1B10 in psoriasis, and explored the potential proteins interacted with circAKR1B10. Levels of genes and proteins were assayed by qRT-PCR and western blotting analyses. Keratinocytes in functional groups were treated with interleukin (IL)-22. Functional analysis were conducted using MTT, 5-ethynyl-2'-deoxyuridine (EdU), and transwell assays, respectively. Interaction analysis among circAKR1B10, Eukaryotic initiation factor 4 A-III (EIF4A3) and Aurora Kinase A (AURKA) was conducted using bioinformatics analysis, RNA pull-down assay, and RNA immunoprecipitation (RIP) assay. CircAKR1B10 was highly expressed in psoriasis patients and IL-22-induced keratinocytes. Functionally, knockdown of circAKR1B10 abolished IL-22-induced proliferation, migration and invasion in keratinocytes. AURKA expression was also higher in psoriasis patients and IL-22-induced keratinocytes, and was negatively correlated with circAKR1B10 expression. Moreover, AURKA silencing reduced the proliferative, migratory and invasive abilities of IL-22-induced keratinocytes. Mechanistically, circAKR1B10 interacted with EIF4A3 protein to stabilize and regulate AURKA expression. CircAKR1B10 contributes to IL-22-induced proliferation, migration and invasion in keratinocytes via up-regulating AURKA expression through interacting with EIF4A3 protein.


Subject(s)
Aurora Kinase A , Cell Movement , Cell Proliferation , Eukaryotic Initiation Factor-4A , Interleukin-22 , Interleukins , Keratinocytes , Psoriasis , RNA, Circular , Humans , Aurora Kinase A/metabolism , Aurora Kinase A/genetics , RNA, Circular/genetics , RNA, Circular/metabolism , Keratinocytes/metabolism , Cell Proliferation/drug effects , Cell Movement/drug effects , Psoriasis/pathology , Psoriasis/metabolism , Psoriasis/genetics , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4A/genetics , Interleukins/metabolism , Interleukins/genetics , DEAD-box RNA Helicases
12.
Sci Immunol ; 9(98): eade7530, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39151019

ABSTRACT

How group 3 innate lymphoid cells (ILC3s) regulate mucosal protection in the presence of T cells remains poorly understood. Here, we examined ILC3 function in intestinal immunity using ILC3-deficient mice that maintain endogenous T cells, T helper 17 (TH17) cells, and secondary lymphoid organs. ILC3s were dispensable for generation of TH17 and TH22 cell responses to commensal and pathogenic bacteria, and absence of ILC3s did not affect IL-22 production by CD4 T cells before or during infection. However, despite the presence of IL-22-producing T cells, ILC3s and ILC3-derived IL-22 were required for maintaining homeostatic functions of the intestinal epithelium. T cell-sufficient, ILC3-deficient mice were capable of pathogen clearance and survived infection with a low dose of Citrobacter rodentium. However, ILC3s promoted pathogen tolerance at early time points of infection by activating tissue-protective immune pathways. Consequently, ILC3s were indispensable for survival after high-dose infection. Our results demonstrate a context-dependent role for ILC3s in immune-sufficient animals and provide a blueprint for uncoupling of ILC3 and TH17 cell functions.


Subject(s)
Citrobacter rodentium , Enterobacteriaceae Infections , Immunity, Innate , Intestinal Mucosa , Lymphocytes , Mice, Inbred C57BL , Animals , Immunity, Innate/immunology , Mice , Lymphocytes/immunology , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice, Knockout , Interleukin-22 , Immunity, Mucosal/immunology , Th17 Cells/immunology
13.
Front Immunol ; 15: 1437046, 2024.
Article in English | MEDLINE | ID: mdl-39156888

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD) comprises a spectrum of liver diseases that span simple steatosis, metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis and may progress to cirrhosis and cancer. The pathogenesis of MASLD is multifactorial and is driven by environmental, genetic, metabolic and immune factors. This review will focus on the role of the type 3 cytokines IL-17 and IL-22 in MASLD pathogenesis and progression. IL-17 and IL-22 are produced by similar adaptive and innate immune cells such as Th17 and innate lymphoid cells, respectively. IL-17-related signaling is upregulated during MASLD resulting in increased chemokines and proinflammatory cytokines in the liver microenvironment, enhanced recruitment of myeloid cells and T cells leading to exacerbation of inflammation and liver disease progression. IL-17 may also act directly by activating hepatic stellate cells resulting in increased fibrosis. In contrast, IL-22 is a pleiotropic cytokine with a dominantly protective signature in MASLD and is currently being tested as a therapeutic strategy. IL-22 also exhibits beneficial metabolic effects and abrogates MASH-related inflammation and fibrosis development via inducing the production of anti-oxidants and anti-apoptotic factors. A sex-dependent effect has been attributed to both cytokines, most importantly to IL-22 in MASLD or related conditions. Altogether, IL-17 and IL-22 are key effectors in MASLD pathogenesis and progression. We will review the role of these two cytokines and cells that produce them in the development of MASLD, their interaction with host factors driving MASLD including sexual dimorphism, and their potential therapeutic benefits.


Subject(s)
Interleukin-17 , Interleukin-22 , Interleukins , Humans , Interleukin-17/metabolism , Interleukin-17/immunology , Interleukins/metabolism , Interleukins/immunology , Animals , Fatty Liver/immunology , Fatty Liver/metabolism , Fatty Liver/pathology , Metabolic Diseases/metabolism , Metabolic Diseases/immunology , Liver/pathology , Liver/metabolism , Liver/immunology
14.
Front Immunol ; 15: 1447431, 2024.
Article in English | MEDLINE | ID: mdl-39211040

ABSTRACT

Mammalian interleukin-22 (IL-22) attenuates organismal injury by inhibiting reactive oxygen species (ROS) and impeding the NLRP3 inflammasome activation. However, the role of fish IL-22 in this process remains unclear. We characterized MaIL-22, an IL-22 homolog in blunt snout bream (Megalobrama amblycephala). Despite its low sequence identity, it shares conserved structures and close evolutionary relationships with other teleost IL-22s. Furthermore, Aeromonas hydrophila (A. hydrophila) infection leads to tissue injury in M. amblycephala immune organs and concomitantly altered Mail-22 mRNA expression, suggesting that MaIL-22 was involved in the antimicrobial immune response. To explore MaIL-22's biological functions, we produced recombinant MaIL-22 (rMaIL-22) protein and demonstrated it significantly enhanced the survival of M. amblycephala post-A. hydrophila infection. To unravel its protective mechanisms, we explored the ROS/NLRP3 inflammasome axis and its downstream signaling responses. The results showed that rMaIL-22 treatment significantly elevated antioxidant enzyme (T-SOD, CAT and GSH-PX) activities to inhibit MDA activity and scavenge ROS in visceral tissues. Meanwhile, rMaIL-22 impeded the activation of NLRP3 inflammasome by suppressing NLRP3 protein and mRNA expression. This indicated that rMaIL-22 contributed to inhibit A. hydrophila-induced activation of the ROS/NLRP3 inflammasome axis. Consistent with these findings, rMaIL-22 treatment attenuated the expression of proinflammatory cytokines (il-1ß, tnf-α and il-6) and proapoptotic genes (caspase-3 and caspase-8) while promoting antiapoptotic genes (bcl-2b and mcl-1a) expression, ultimately mitigating tissue injury in visceral tissues. In conclusion, our research underscores MaIL-22's key role in microbial immune regulation, offering insights for developing IL-22-targeted therapies and breeding programs.


Subject(s)
Aeromonas hydrophila , Apoptosis , Fish Diseases , Gram-Negative Bacterial Infections , Inflammasomes , Inflammation , Interleukin-22 , Interleukins , NLR Family, Pyrin Domain-Containing 3 Protein , Reactive Oxygen Species , Animals , Aeromonas hydrophila/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Inflammasomes/immunology , Apoptosis/drug effects , Gram-Negative Bacterial Infections/immunology , Reactive Oxygen Species/metabolism , Interleukins/metabolism , Interleukins/immunology , Fish Diseases/immunology , Fish Diseases/microbiology , Inflammation/immunology , Fish Proteins/metabolism , Fish Proteins/immunology , Fish Proteins/genetics , Cyprinidae/immunology , Cyprinidae/metabolism , Signal Transduction
15.
Commun Biol ; 7(1): 804, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961129

ABSTRACT

Oral administration of harmless antigens can induce suppression of reactive immune responses, a process that capitalises on the ability of the gastrointestinal tract to tolerate exposure to food and commensal microbiome without triggering inflammatory responses. Repeating exposure to type II collagen induces oral tolerance and inhibits induction of arthritis, a chronic inflammatory joint condition. Although some mechanisms underlying oral tolerance are described, how dysregulation of gut immune networks impacts on inflammation of distant tissues like the joints is unclear. We used undenatured type II collagen in a prophylactic regime -7.33 mg/kg three times/week- to describe the mechanisms associated with protective oral immune-therapy (OIT) in gut and joint during experimental Collagen-Induced Arthritis (CIA). OIT reduced disease incidence to 50%, with reduced expression of IL-17 and IL-22 in the joints of asymptomatic mice. Moreover, whilst the gut tissue of arthritic mice shows substantial damage and activation of tissue-specific immune networks, oral administration of undenatured type II collagen protects against gut pathology in all mice, symptomatic and asymptomatic, rewiring IL-17/IL-22 networks. Furthermore, gut fucosylation and microbiome composition were also modulated. These results corroborate the relevance of the gut-joint axis in arthritis, showing novel regulatory mechanisms linked to therapeutic OIT in joint disease.


Subject(s)
Arthritis, Experimental , Collagen Type II , Gastrointestinal Microbiome , Homeostasis , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/prevention & control , Collagen Type II/immunology , Mice , Gastrointestinal Microbiome/drug effects , Male , Joints/immunology , Joints/drug effects , Joints/pathology , Mice, Inbred DBA , Interleukin-17/metabolism , Interleukin-22 , Administration, Oral
16.
J Exp Med ; 221(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38949650

ABSTRACT

Germline activating mutations in STAT3 cause a multi-systemic autoimmune and autoinflammatory condition. By studying a mouse model, Toth et al. (https://doi.org/10.1084/jem.20232091) propose a role for dysregulated IL-22 production by Th17 cells in causing some aspects of immune-mediated skin inflammation in human STAT3 GOF syndrome.


Subject(s)
Interleukin-22 , STAT3 Transcription Factor , Skin , Th17 Cells , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Animals , Humans , Th17 Cells/immunology , Th17 Cells/metabolism , Skin/metabolism , Skin/pathology , Interleukins/genetics , Interleukins/metabolism , Gain of Function Mutation , Mice , Inflammation/metabolism
17.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 744-748, 2024 May 20.
Article in Chinese | MEDLINE | ID: mdl-38948276

ABSTRACT

Objective: To investigate the roles of histone H3K27me3 methylation and its regulatory enzymes JMJD3 and EZH2 in the differentiation of Th17 cells in ankylosing spondylitis (AS), to unveil their potential involvement in the pathogenesis of AS, and to provide new strategies and targets for the clinical treatment of AS by analyzing the methylation state of H3K27me3 and its interactions with Th17-related factors. Methods: A total of 84 AS patients (42 active AS patiens and 42 patients in the stable phase of AS) were enrolled for the study, while 84 healthy volunteers were enrolled as the controls. Blood samples were collected. Peripheral blood mononuclear cells were isolated. ELISA assay was performed to examine Th17 cells and the relevant cytokines IL-21, IL-22, and IL-17. The mRNA expressions of RORc, JAK2, and STAT3 were analyzed by RT-PCR, the protein expressions of RORc, JAK2/STAT3 pathway protein, H3K27me3 and the relevant protease (EZH2 and JMJD3) were determined by Western blot. Correlation between H3K27me3, EZH2 and JMJD3 and the key signaling pathway molecules of Th cell differentiation was analyzed by Pearson correlation analysis. Results: The mRNA expressions of RORc, JAK2, and STAT3 were significantly higher in the active phase group than those in the stable phase group ( P<0.05). The relative grayscale values of H3K27me3 and EZH2 in the active phase group were lower than those of the stable phase group, which were lower than those of the control group, with the differences being statistically significant ( P<0.05). The relative grayscale values of JMJD3, RORc, JAK2, pJAK2, STAT3, and pSTAT3 proteins were significantly higher in the active phase group than those in the stable phase group, which were higher than those in the control group (all P<0.05). The proportion of Th17 and the expression level of inflammatory factors in the active period group were higher than those in the other two groups (P<0.05). H3K27me3 was negatively correlated with RORc, JAK2, STAT3, and IL-17, JMJD3 was positvely correlated with JAK2, STAT3, and IL-17, and EZH2 was negatively correlated with JAK2, STAT3, and IL-17 (all P<0.05). Conclusion: The low expression of H3K27me3 in AS is influenced by the gene loci JMJD3 and EZH2, which can regulate the differentiation of Th17 cells and thus play a role in the pathogenesis and progression of AS.


Subject(s)
Cell Differentiation , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Histones , Interleukin-17 , Jumonji Domain-Containing Histone Demethylases , Nuclear Receptor Subfamily 1, Group F, Member 3 , STAT3 Transcription Factor , Spondylitis, Ankylosing , Th17 Cells , Humans , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/metabolism , Th17 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Histones/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Interleukin-17/metabolism , Interleukin-17/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Methylation , Interleukins/metabolism , Interleukins/genetics , Interleukin-22 , Male , Female , Adult
18.
J Neuroimmune Pharmacol ; 19(1): 37, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052165

ABSTRACT

Recombinant interleukin-22 (rIL-22) has been reported as a protective agent in murine models of diseases driven by epithelial injury. Parasites have a circadian rhythm and their sensitivity to a certain drug may vary during the day. Therefore, this work aimed to investigate the effect of rIL-22 administration at different times of the day on the inflammation, oxidative status, and neurotransmitter release in the gut-brain axis of the Schistosoma mansoni-infected mice. Sixty male BALB/c mice aged six weeks weighing 25-30 g were divided into a control group (injected intraperitoneally with PBS), mice infected with 80 ± 10 cercariae of S. mansoni (infected group) then injected intraperitoneally with PBS, and rIL-22 treated groups. rIL-22 was administrated intraperitoneally (400 ng/kg) either at the onset or offset of the light phase for 14 days. IL-22 administration reduced the levels of IL-1ß, tumor necrosis factor-alpha (TNF-α), nuclear factor kappa beta (NF-κß), and enhanced the production of IL-22 and IL-17. The treatment with IL-22 increased glutathione (GSH) and reduced malondialdehyde (MDA) and nitric oxide (NO) levels both in the ileum and brain. The B-cell lymphoma 2 (BCL2) protein level in the ileum was diminished after IL-22 administration. Brain-derived neurotrophic factor (BDNF) and neurotransmitter release (serotonin, 5HT, norepinephrine, NE, dopamine, DA, Glutamate, Glu, and -amino butyric acid, GABA) were improved by rIL-22. In conclusion, rIL-22 showed promising immunotherapy for inflammation, oxidative damage, and neuropathological signs associated with schistosomiasis. The efficacy of IL-22 increased significantly upon its administration at the time of light offset.


Subject(s)
Brain-Gut Axis , Interleukin-22 , Interleukins , Mice, Inbred BALB C , Neurotransmitter Agents , Recombinant Proteins , Schistosomiasis mansoni , Animals , Mice , Male , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/pharmacology , Interleukins/metabolism , Schistosomiasis mansoni/drug therapy , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/administration & dosage , Brain-Gut Axis/drug effects , Brain-Gut Axis/physiology , Immunotherapy/methods , Biogenic Monoamines/metabolism , Inflammation/metabolism , Inflammation/drug therapy
19.
Nature ; 631(8022): 857-866, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987586

ABSTRACT

Systemic lupus erythematosus (SLE) is prototypical autoimmune disease driven by pathological T cell-B cell interactions1,2. Expansion of T follicular helper (TFH) and T peripheral helper (TPH) cells, two T cell populations that provide help to B cells, is a prominent feature of SLE3,4. Human TFH and TPH cells characteristically produce high levels of the B cell chemoattractant CXCL13 (refs. 5,6), yet regulation of T cell CXCL13 production and the relationship between CXCL13+ T cells and other T cell states remains unclear. Here, we identify an imbalance in CD4+ T cell phenotypes in patients with SLE, with expansion of PD-1+/ICOS+ CXCL13+ T cells and reduction of CD96hi IL-22+ T cells. Using CRISPR screens, we identify the aryl hydrocarbon receptor (AHR) as a potent negative regulator of CXCL13 production by human CD4+ T cells. Transcriptomic, epigenetic and functional studies demonstrate that AHR coordinates with AP-1 family member JUN to prevent CXCL13+ TPH/TFH cell differentiation and promote an IL-22+ phenotype. Type I interferon, a pathogenic driver of SLE7, opposes AHR and JUN to promote T cell production of CXCL13. These results place CXCL13+ TPH/TFH cells on a polarization axis opposite from T helper 22 (TH22) cells and reveal AHR, JUN and interferon as key regulators of these divergent T cell states.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , CD4-Positive T-Lymphocytes , Chemokine CXCL13 , Interferon Type I , Lupus Erythematosus, Systemic , Proto-Oncogene Proteins c-jun , Receptors, Aryl Hydrocarbon , Female , Humans , Male , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Chemokine CXCL13/metabolism , Epigenomics , Gene Expression Profiling , Interferon Type I/immunology , Interferon Type I/metabolism , Interleukin-22/immunology , Interleukin-22/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/genetics , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Aryl Hydrocarbon/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
20.
Respir Res ; 25(1): 275, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003443

ABSTRACT

Pleurisy can be categorized as primary or secondary, arising from immunological, tumorous, or microbial conditions. It often results in lung structure damage and the development of various respiratory issues. Among the different types, tuberculous pleurisy has emerged as a prominent focus for both clinical and scientific investigations. The IL-10 family, known for its anti-inflammatory properties in the human immune system, is increasingly being studied for its involvement in the pathogenesis of pleurisy. This review aims to present a detailed overview of the intricate role of IL-10 family members (specifically IL-10, IL-22, and IL-26) in human and animal pleuritic diseases or relevant animal models. These insights could serve as valuable guidance and references for further studies on pleurisy and potential therapeutic strategies.


Subject(s)
Interleukin-10 , Interleukin-22 , Interleukins , Tuberculosis, Pleural , Animals , Humans , Interleukin-10/metabolism , Interleukins/metabolism , Interleukins/immunology , Pleurisy/immunology , Pleurisy/diagnosis , Pleurisy/metabolism , Tuberculosis, Pleural/diagnosis , Tuberculosis, Pleural/immunology , Tuberculosis, Pleural/metabolism , Tuberculosis, Pleural/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL