Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 245
Filter
Add more filters

Publication year range
1.
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
2.
J Nutr Biochem ; 101: 108926, 2022 03.
Article in English | MEDLINE | ID: mdl-34848335

ABSTRACT

Short-chain fatty acids (SCFAs) play a pivotal role in maintaining intestinal homeostasis. We aimed to investigate the effects of SCFA supplementation on gut inflammation and microbiota composition in a murine colitis model. Mice were fed with sodium butyrate or a mixture of SCFAs in the drinking water for 2 weeks, followed by 2% dextran sulfate sodium (DSS) for 7 d. After euthanasia, mouse colons were extracted to examine histological findings. Flow cytometry of the mouse colon tissues was performed to assess T cell differentiation. Changes in gut microbiota were assessed by high-throughput sequencing of the mouse feces. There were no significant differences in weight change, colonic length, or histologic inflammation score between the DSS, butyrate, and SCFA mix groups. However, flow cytometry revealed that both the expression of CD4+Foxp3+ regulatory T cells and of IL-17-producing T cells were increased in the butyrate and SCFA mix groups. Microbial compositions of the butyrate and SCFA mix groups were significantly different from those of the control and DSS groups in principal coordinate analysis. Relative abundances of the phyla Verrucomicrobia and Proteobacteria, species Akkermansia muciniphila and Escherichia fergusonii were increased in the butyrate and SCFA mix groups. Genera Roseburia and Lactobacillus showed a negative correlation with the degree of colitis, whereas genera Escherichia and Mucispirillum showed a positive correlation. SCFA supplementation did not result in a significant reduction in colon inflammation, but it promoted both regulatory T cell and IL-17-producing T cell expression, and increased both protective and aggressive gut microbiota.


Subject(s)
Butyrates/administration & dosage , Dietary Supplements , Fatty Acids, Volatile/administration & dosage , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Animals , Cell Differentiation , Colitis/immunology , Colitis/microbiology , Colitis/pathology , Colon/pathology , Disease Models, Animal , Feces/microbiology , Female , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Inflammatory Bowel Diseases/immunology , Mice , Mice, Inbred C57BL , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology
3.
Int J Biol Macromol ; 195: 142-151, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34896465

ABSTRACT

In the present research, the water-soluble polysaccharides (AMP) from Atractylodes macrocephalae Koidz. were isolated and prepared. The protective effects of AMP on intestinal mucosal barrier injury induced by dextran sulfate sodium (DSS) in mice were investigated. It was found that AMP treatment significantly alleviated the body weight decreases and shorten colon length, and ameliorated colonic damage induced by DSS. Importantly, AMP prevented the over-expression of proinflammatory cytokines TNF-α, IL-1ß and IL-6, and decreased the infiltration of neutrophils in colon. Additionally, AMP could raise expressions of Mucin 2 and tight junction protein Claudin-1. AMP also modulated the intestinal microbiota by enhancing the overall richness and diversity, greatly reducing the proportion of harmful bacteria, for instance, Clostridiumsensu stricto1 and Escherichia Shigella, however, augmenting the ratio of potential beneficial bacteria such as Faecalibaculum and Bifidobacterium. This work offers some important insights on protective effects of polysaccharides AMP against intestinal barrier dysfunction and provides underlying mechanism of health-beneficial properties of these biological macromolecules.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Atractylodes/chemistry , Colitis/drug therapy , Dextran Sulfate/adverse effects , Intestinal Mucosa/injuries , Polysaccharides/administration & dosage , Animals , Anti-Inflammatory Agents/pharmacology , Bacteria/classification , Bacteria/drug effects , Bacteria/isolation & purification , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Intestinal Mucosa/drug effects , Male , Mice , Phylogeny , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
4.
J Nutr Biochem ; 99: 108855, 2022 01.
Article in English | MEDLINE | ID: mdl-34517096

ABSTRACT

Patients with inflammatory bowel disease (IBD) have higher incidence of extraintestinal manifestations (EIM), including liver disorders, sarcopenia, and neuroinflammation. Fermented rice bran (FRB), generated from rice bran (RB), is rich in bioactive compounds, and exhibits anti-colitis activity. However, its role in EIM prevention is still unclear. Here, for the first time, we investigated whether EIM in female C57Bl/6N mice is attenuated by FRB supplementation. EIM was induced by repeated administration of 1.5% dextran sulfate sodium (DSS) in drinking water (4 d) followed by drinking water (12 d). Mice were divided into 3 groups-control (AIN93M), 10% RB, and 10% FRB. FRB ameliorated relapsing colitis and inflammation in muscle by significantly lowering proinflammatory cytokines Tnf-α and Il-6 in serum and advanced glycation end product-specific receptor (Ager) in serum and muscle when compared with the RB and control groups. As FRB reduced aspartate aminotransferase levels and oxidative stress, it might prevent liver disorders. FRB downregulated proinflammatory cytokine and chemokine transcripts responsible for neuroinflammation in the hippocampus and upregulated mRNA expression of G protein coupled receptors (GPRs), Gpr41 and Gpr43, in small and large intestines, which may explain the FRB-mediated protective mechanism. Hence, FRB can be used as a supplement to prevent IBD-associated EIM.


Subject(s)
Colitis/drug therapy , Colitis/immunology , Dietary Fiber/administration & dosage , Oryza/chemistry , Plant Preparations/administration & dosage , Animals , Chemokines/genetics , Chemokines/immunology , Chronic Disease/therapy , Colitis/chemically induced , Colitis/genetics , Dextran Sulfate/adverse effects , Dietary Fiber/analysis , Dietary Supplements/analysis , Disease Models, Animal , Female , Hippocampus/immunology , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Intestines/immunology , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/immunology , Oxidative Stress , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
5.
Cell Host Microbe ; 30(1): 17-30.e9, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34822777

ABSTRACT

Bacterial activation of T helper 17 (Th17) cells exacerbates mouse models of autoimmunity, but how human-associated bacteria impact Th17-driven disease remains elusive. We show that human gut Actinobacterium Eggerthella lenta induces intestinal Th17 activation by lifting inhibition of the Th17 transcription factor Rorγt through cell- and antigen-independent mechanisms. E. lenta is enriched in inflammatory bowel disease (IBD) patients and worsens colitis in a Rorc-dependent manner in mice. Th17 activation varies across E. lenta strains, which is attributable to the cardiac glycoside reductase 2 (Cgr2) enzyme. Cgr2 is sufficient to induce interleukin (IL)-17a, a major Th17 cytokine. cgr2+ E. lenta deplete putative steroidal glycosides in pure culture; related compounds are negatively associated with human IBD severity. Finally, leveraging the sensitivity of Cgr2 to dietary arginine, we prevented E. lenta-induced intestinal inflammation in mice. Together, these results support a role for human gut bacterial metabolism in driving Th17-dependent autoimmunity.


Subject(s)
Colitis/metabolism , Gastrointestinal Microbiome/physiology , Lymphocyte Activation/physiology , Th17 Cells/metabolism , Actinobacteria , Animals , Bacteria/metabolism , Colitis/immunology , Cytokines , Dietary Supplements , Disease Models, Animal , Female , Humans , Inflammatory Bowel Diseases/microbiology , Interleukin-17/metabolism , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
6.
Front Immunol ; 12: 753092, 2021.
Article in English | MEDLINE | ID: mdl-34745126

ABSTRACT

Increasing evidence support that cellular amino acid metabolism shapes the fate of immune cells; however, whether aspartate metabolism dictates macrophage function is still enigmatic. Here, we found that the metabolites in aspartate metabolism are depleted in lipopolysaccharide (LPS) plus interferon gamma (IFN-γ)-stimulated macrophages. Aspartate promotes interleukin-1ß (IL-1ß) secretion in M1 macrophages. Mechanistically, aspartate boosts the activation of hypoxia-inducible factor-1α (HIF-1α) and inflammasome and increases the levels of metabolites in aspartate metabolism, such as asparagine. Interestingly, asparagine also accelerates the activation of cellular signaling pathways and promotes the production of inflammatory cytokines from macrophages. Moreover, aspartate supplementation augments the macrophage-mediated inflammatory responses in mice and piglets. These results uncover a previously uncharacterized role for aspartate metabolism in directing M1 macrophage polarization.


Subject(s)
Aspartic Acid/metabolism , Inflammasomes/physiology , Interleukin-1beta/biosynthesis , Macrophages, Peritoneal/immunology , Animals , Citrobacter rodentium , Colitis/immunology , Colitis/microbiology , Cytokines/blood , Enterobacteriaceae Infections/immunology , Female , Hypoxia-Inducible Factor 1, alpha Subunit , Interferon-gamma/pharmacology , Interleukin-1beta/genetics , Lipopolysaccharides/pharmacology , Macrophage Activation , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Swine
7.
Front Immunol ; 12: 762564, 2021.
Article in English | MEDLINE | ID: mdl-34675940

ABSTRACT

Accumulating evidences support that amino acids direct the fate decision of immune cells. Glycine is a simple structural amino acid acting as an inhibitory neurotransmitter. Besides, glycine receptors as well as glycine transporters are found in macrophages, indicating that glycine alters the functions of macrophages besides as an inhibitory neurotransmitter. Mechanistically, glycine shapes macrophage polarization via cellular signaling pathways (e.g., NF-κB, NRF2, and Akt) and microRNAs. Moreover, glycine has beneficial effects in preventing and/or treating macrophage-associated diseases such as colitis, NAFLD and ischemia-reperfusion injury. Collectively, this review highlights the conceivable role of glycinergic signaling for macrophage polarization and indicates the potential application of glycine supplementation as an adjuvant therapy in macrophage-associated diseases.


Subject(s)
Glycine/immunology , Macrophages/immunology , Animals , Colitis/immunology , Glycine/metabolism , Humans , Metabolic Diseases/immunology , MicroRNAs , Neoplasms/immunology , Reperfusion Injury/immunology , Signal Transduction
8.
Nat Immunol ; 22(11): 1440-1451, 2021 11.
Article in English | MEDLINE | ID: mdl-34686860

ABSTRACT

Intestinal epithelial cell (IEC) damage by T cells contributes to graft-versus-host disease, inflammatory bowel disease and immune checkpoint blockade-mediated colitis. But little is known about the target cell-intrinsic features that affect disease severity. Here we identified disruption of oxidative phosphorylation and an increase in succinate levels in the IECs from several distinct in vivo models of T cell-mediated colitis. Metabolic flux studies, complemented by imaging and protein analyses, identified disruption of IEC-intrinsic succinate dehydrogenase A (SDHA), a component of mitochondrial complex II, in causing these metabolic alterations. The relevance of IEC-intrinsic SDHA in mediating disease severity was confirmed by complementary chemical and genetic experimental approaches and validated in human clinical samples. These data identify a critical role for the alteration of the IEC-specific mitochondrial complex II component SDHA in the regulation of the severity of T cell-mediated intestinal diseases.


Subject(s)
Colitis/enzymology , Colon/enzymology , Cytotoxicity, Immunologic , Electron Transport Complex II/metabolism , Epithelial Cells/enzymology , Graft vs Host Disease/enzymology , Intestinal Mucosa/enzymology , Mitochondria/enzymology , T-Lymphocytes/immunology , Animals , Case-Control Studies , Cell Communication , Cells, Cultured , Colitis/genetics , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/ultrastructure , Disease Models, Animal , Electron Transport Complex II/genetics , Epithelial Cells/immunology , Epithelial Cells/ultrastructure , Female , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/ultrastructure , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/immunology , Mitochondria/ultrastructure , Oxidative Phosphorylation , Succinic Acid/metabolism , T-Lymphocytes/metabolism
9.
Int Immunopharmacol ; 101(Pt A): 108191, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34601328

ABSTRACT

Neobavaisoflavone (Neo), is the active constituent of the herb Psoralea corylifolial, used in the traditional Chinese medicine, and has anti-inflammatory activity, but whether Neo could regulate colitis remains unclear. T helper 9 (TH9) cells, a subset of CD4+ T helper cells characterized by secretion of IL-9, have been reported to be involved in the pathogenesis of many autoimmune and inflammatory diseases, but whether Neo could control TH9 cell differentiation also remains unclear. Here, we found that Neo could decrease IL-9 production of CD4+ T cells by targeting PU.1 in vitro. Importantly, Neo had therapeutic effects on DSS-induced colitis. Furthermore, we identified TH9 cells as the direct target of Neo for attenuating bowel inflammation. Therefore, Neo could serve as a lead for developing new therapeutics against inflammatory bowel disease.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cell Differentiation/drug effects , Colitis/drug therapy , Isoflavones/therapeutic use , T-Lymphocytes, Helper-Inducer/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Colitis/immunology , Female , Fluorescent Antibody Technique , Humans , Immunoblotting , Isoflavones/pharmacology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Real-Time Polymerase Chain Reaction
10.
Biomed Pharmacother ; 143: 112148, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34560553

ABSTRACT

Ulcerative colitis (UC) is a gastrointestinal inflammatory disease with a multifactorial pathophysiology. This study aims to investigate the immunomodulatory effect of Portulaca oleracea leaf ethanolic extract (POE) on acetic acid (AA)-induced UC in mice. Experimental animals received oral doses of POE (200 mg/kg for 7 days) after an induction of colitis by intrarectal AA administration. In mice with AA-induced UC treated with POE, the results revealed a significant modulation in body weight and colon length. Moreover, treatment with POE downregulated the interleukin 1, 6, and 17, tumor necrosis factor-alpha, gamma interferon, and nuclear factor-kappa B levels compared with the colitis group. Furthermore, POE markedly inhibited histological damage, decreased myeloperoxidase activity and reduced fecal calprotectin level compared with the colitis group. These data are consistent with the reduction in total bacterial content in the colon. Taken together, treatment with POE may reduce colonic inflammation by alleviating the immune response and inhibiting the severity of colitis. The HPLC analysis of POE resulted in the identification of seven medicinal compounds comprising two phenolic acids (ferulic and caffeic acids) and five flavonoids (kaempferol, quercetin, rutin, narenginin and hesperidin). Subsequent analysis of POE by GC-MS revealed ten phytocomponents; the major percentages were hexadecenoic acid, methyl ester (29.8119%), α-linolenic acid (25.8431%), 16-octadecenoic acid, methyl ester (15.1578%) and α-tocopherol (10.7848%). Delta-lactams and alkanes were the minor components. Such natural plant-derived substances and their probable synergistic action appear to contribute to a promising therapeutic protocol for colitis.


Subject(s)
Colitis/drug therapy , Immunomodulating Agents/pharmacology , Phytochemicals/pharmacology , Plant Extracts/pharmacology , Portulaca , Animals , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , Cytokines/metabolism , Disease Models, Animal , Gastrointestinal Microbiome , Immunomodulating Agents/isolation & purification , Inflammation Mediators/metabolism , Leukocyte L1 Antigen Complex/metabolism , Male , Mice , NF-kappa B/metabolism , Peroxidase/metabolism , Phytochemicals/isolation & purification , Plant Extracts/isolation & purification , Plant Leaves , Portulaca/chemistry
11.
J Immunol Res ; 2021: 9117805, 2021.
Article in English | MEDLINE | ID: mdl-34195297

ABSTRACT

Although the strain-dependent effects of Bacteroides vulgatus on alleviating intestinal inflammatory diseases have been demonstrated, the literature has rarely focused on the underlying causes of this effect. In this study, we selected four B. vulgatus strains (FTJS5K1, FTJS7K1, FSDTA11B14, and FSDLZ51K1) with different genomic characteristics and evaluated their protective roles against dextran sulfate sodium- (DSS-) induced colitis. Compared to the other three tested strains, B. vulgatus 7K1 more strongly ameliorated the DSS-induced weight loss, shortening of the colon length, increased disease activity index scores, colonic tissue injury, and immunomodulatory disorder. In contrast, B. vulgatus 51K1 significantly worsened the DSS-induced alterations in the tumor necrosis factor-alpha (TNF-α) concentration and colonic histopathology. A comparative genomic analysis of B. vulgatus 7K1 and 51K1 showed that the beneficial effects of B. vulgatus 7K1 may be associated with some of its specific genes involved in the production of short-chain fatty acids or capsular polysaccharides and enhancement of its survivability in the gut. In conclusion, these findings indicate that the supplementation of B. vulgatus 7K1 is a potentially efficacious intervention for alleviating colitis and provides scientific support for the screening of probiotics with anticolitis effect.


Subject(s)
Bacteroides/genetics , Colitis/microbiology , Colon/pathology , Inflammatory Bowel Diseases/microbiology , Animals , Colitis/chemically induced , Colitis/immunology , Dextran Sulfate , Disease Models, Animal , Fatty Acids/metabolism , Humans , Inflammatory Bowel Diseases/immunology , Male , Mice , Mice, Inbred C57BL , Polysaccharides/metabolism , Species Specificity , Tumor Necrosis Factor-alpha/metabolism
12.
Molecules ; 26(11)2021 Jun 02.
Article in English | MEDLINE | ID: mdl-34199466

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal tract with an incompletely understood pathogenesis. Long-standing colitis is associated with increased risk of colon cancer. Despite the availability of various anti-inflammatory and immunomodulatory drugs, many patients fail to respond to pharmacologic therapy and some experience drug-induced adverse events. Dietary supplements, particularly saffron (Crocus sativus), have recently gained an appreciable attention in alleviating some symptoms of digestive diseases. In our study, we investigated whether saffron may have a prophylactic effect in a murine colitis model. Saffron pre-treatment improved the gross and histopathological characteristics of the colonic mucosa in murine experimental colitis. Treatment with saffron showed a significant amelioration of colitis when compared to the vehicle-treated mice group. Saffron treatment significantly decreased secretion of serotonin and pro-inflammatory cytokines, such as TNF-α, IL-1ß, and IL-6, in the colon tissues by suppressing the nuclear translocation of NF-κB. The gut microbiome analysis revealed distinct clusters in the saffron-treated and untreated mice in dextran sulfate sodium (DSS)-induced colitis by visualization of the Bray-Curtis diversity by principal coordinates analysis (PCoA). Furthermore, we observed that, at the operational taxonomic unit (OTU) level, Cyanobacteria were depleted, while short-chain fatty acids (SCFAs), such as isobutyric acid, acetic acid, and propionic acid, were increased in saffron-treated mice. Our data suggest that pre-treatment with saffron inhibits DSS-induced pro-inflammatory cytokine secretion, modulates gut microbiota composition, prevents the depletion of SCFAs, and reduces the susceptibility to colitis.


Subject(s)
Bacteria/classification , Biological Products/administration & dosage , Colitis/drug therapy , Crocus/chemistry , Dextran Sulfate/adverse effects , Microbiota/drug effects , Administration, Oral , Animals , Bacteria/drug effects , Bacteria/genetics , Bacteria/isolation & purification , Biological Products/pharmacology , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Cytokines/metabolism , Disease Models, Animal , Down-Regulation , Male , Mice , Mice, Inbred C57BL , Phylogeny , Pre-Exposure Prophylaxis , Serotonin/metabolism , Treatment Outcome
13.
J Allergy Clin Immunol ; 148(4): 1081-1087.e2, 2021 10.
Article in English | MEDLINE | ID: mdl-34019913

ABSTRACT

BACKGROUND: CD56-expressing natural killer (NK) cells as well as invariant NK T (iNKT) cells have been shown to either promote or inhibit allergic immune responses. OBJECTIVE: The aim of the present study was to investigate the impact of these cells in a recently developed humanized mouse model of allergen-induced IgE-dependent gut and lung inflammation. METHODS: Nonobese diabetic-severe combined immunodeficiency γ-chain knockout mice were injected intraperitoneally with human PBMCs or CD56-depleted (CD56neg) PBMCs from highly sensitized donors with birch or grass pollen allergy together with the respective allergen or with NaCl as a control. Three weeks later, the mice were challenged with the allergen rectally and gut inflammation was monitored by video miniendoscopy and by histology. Furthermore, airway inflammation was measured after an additional intranasal allergen challenge. RESULTS: Allergen-specific human IgE in mouse sera, detectable only after coinjection of the respective allergen, was reduced in mice being injected with CD56neg PBMCs compared with in mice receiving nondepleted PBMCs. Consequently, allergen-induced IgE-dependent colitis, airway hyperreactivity, and mucus-producing goblet cells were significantly inhibited in these mice. Interestingly, reconstitution of CD56neg PBMCs with nondepleted CD56+ cells and with CD56+CD3+ iNKT cells restored gut as well as lung inflammation, whereas addition of CD3-depleted CD56+ cells did not. CONCLUSION: These results demonstrate that allergen-specific gut and lung inflammation in PBMC-engrafted humanized mice is promoted by CD56+CD3+ iNKT cells, which opens new possibilities of therapeutic intervention in allergic diseases.


Subject(s)
Colitis/immunology , Natural Killer T-Cells/immunology , Respiratory Hypersensitivity/immunology , Rhinitis, Allergic, Seasonal/immunology , Allergens/immunology , Animals , Betula/immunology , CD3 Complex/immunology , CD56 Antigen/immunology , Colitis/pathology , Colitis/physiopathology , Colon/immunology , Colon/pathology , Female , Humans , Immunoglobulin E/blood , Lung/immunology , Lung/pathology , Lung/physiopathology , Male , Mice, Transgenic , Poaceae/immunology , Pollen/immunology , Respiratory Hypersensitivity/pathology , Respiratory Hypersensitivity/physiopathology , Rhinitis, Allergic, Seasonal/pathology , Rhinitis, Allergic, Seasonal/physiopathology
14.
Food Funct ; 12(9): 3831-3841, 2021 May 11.
Article in English | MEDLINE | ID: mdl-33977958

ABSTRACT

The aim of the present study was to determine the inhibitory effects and the potential underlying mechanisms of a novel Pleurotus eryngii ß-type glycosidic polysaccharide (WPEP) on colitis. To achieve this, sixty CD-1 (ICR) mice were divided into six groups including healthy and colitic mice treated with or without WPEP at two different doses (n = 10). The results showed that WPEP displayed a significant inhibitory effect on colitis as indicated by the lowered disease activity index in the treated colitic mice compared to the untreated colitic mice (2.78 ± 0.50 to 1.80 ± 0.17). A decrease in pro-inflammatory cytokine concentrations and pro-inflammatory protein expressions and an increase in the colon length (9.31 ± 0.59 cm to 10.89 ± 1.20 cm) along with histological improvements were also observed in the treated colitic mice compared to the untreated colitic mice in the present study. Flow cytometry and western blotting analysis revealed that these anti-colitis effects were associated with decreased accumulation of CD45+ immune cells, CD45 + F4/80+ macrophages and CD45 + Gr1+ neutrophils. Moreover, the 16s rRNA sequencing analysis of the gut microbiota revealed that WPEP partially reversed gut microbiota dysbiosis in the colitic mice including the decreased abundance of Akkermansia muciniphila (35.80 ± 9.10% to 18.24 ± 6.23%) and Clostridium cocleatum (2.34 ± 1.78% to 0.011 ± 0.003%) and the increased abundance of Bifidobacterium pseudolongum (3.48 ± 2.72% to 9.65 ± 3.74%), Lactobacillus reuteri (0.007 ± 0.002% to 0.21 ± 0.12%), Lactobacillus salivarius (1.23 ± 0.87% to 2.22 ± 1.53%) and Ruminococcus bromii (0.009 ± 0.001% to 3.83 ± 1.98%). In summary, our results demonstrated that WPEP could be utilized as a functional food component in colitis management as well as a potential prebiotic agent to improve inflammation-related disorders.


Subject(s)
Colitis/diet therapy , Colon , Dietary Supplements , Glycosides/administration & dosage , Pleurotus/chemistry , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Gastrointestinal Microbiome , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred ICR , Proteins/metabolism
15.
Am J Chin Med ; 49(4): 941-963, 2021.
Article in English | MEDLINE | ID: mdl-33827384

ABSTRACT

Artemisia gmelinii Web. ex Stechm. (AG), a popular medicinal herb in Asia, has been used as a common food ingredient in Korea and is traditionally known for its anti-inflammatory properties. Therefore, in this study, we aimed to investigate whether AG relieves IBD, a classic chronic inflammatory disease of the gastrointestinal tract. We identified 35 chemical compounds in AG ethanol extract using ultra-high-performance liquid chromatography-quadrupole time-of-flight tandem mass spectrometry. In mice with DSS-induced IBD, AG administration attenuated the disease activity index and the serum and colonic levels of inflammatory cytokines and chemokines. AG treatment decreased nuclear factor-[Formula: see text]B (NF-[Formula: see text]B) signaling, a key mediator of inflammation, in the mouse colons. Additionally, AG extract enhanced immune responses in lymphoid tissues such as spleen and Peyer's patches. Thus, AG consumption potently ameliorated IBD symptoms and improved immune signaling in lymphoid tissues.


Subject(s)
Artemisia/chemistry , Colitis/drug therapy , Colitis/immunology , Immunity/drug effects , Inflammation/drug therapy , Plant Extracts/chemistry , Plant Extracts/pharmacology , Animals , Chromatography, High Pressure Liquid , Dextran Sulfate , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Republic of Korea , Tandem Mass Spectrometry
16.
Molecules ; 26(6)2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33808686

ABSTRACT

The incidence and prevalence of inflammatory bowel disorders (IBD) are increasing around the world due to bacterial infection, abnormal immune response, etc. The conventional medicines for IBD treatment possess serious side effects. Periplaneta americana (P. americana), a traditional Chinese medicine, has been used to treat arthritis, fever, aches, inflammation, and other diseases. This study aimed to evaluate the anti-inflammatory effects of oligosaccharides from P. Americana (OPA) and its possible mechanisms in vivo. OPA were purified and biochemical characterization was analyzed by HPGPC, HPLC, FT-IR, and GC-MS. Acute colitis mice model was established, the acute toxicity and anti-inflammatory activity were tested in vivo. The results showed OPA with molecular mass of 1.0 kDa were composed of 83% glucose, 6% galactose, 11% xylose, and the backbone was (1→4)-Glcp. OPA had potent antioxidant activities in vitro and significantly alleviated the clinical symptoms of colitis, relieved colon damage without toxic side effects in vivo. OPA exhibited anti-inflammatory activity by regulating Th1/Th2, reducing oxidative stress, preserving intestinal barrier integrity, and inhibiting TLR4/MAPK/NF-κB pathway. Moreover, OPA protected gut by increasing microbial diversity and beneficial bacteria, and reducing pathogenic bacteria in feces. OPA might be the candidate of complementary and alternative medicines of IBD with low-cost and high safety.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis , Gastrointestinal Microbiome/drug effects , Immunomodulation/drug effects , Oligosaccharides/pharmacology , Periplaneta/chemistry , Acute Disease , Animals , Anti-Inflammatory Agents/chemistry , Colitis/drug therapy , Colitis/immunology , Colitis/microbiology , Disease Models, Animal , Male , Mice , Oligosaccharides/chemistry
17.
Front Immunol ; 12: 648162, 2021.
Article in English | MEDLINE | ID: mdl-33868283

ABSTRACT

Tremella fuciformis is an edible medicinal mushroom, and its polysaccharide components are found to confer various health benefits. This study identified the protective effects of polysaccharides of Tremella fuciformis (TPs) against dextran sulfate sodium (DSS)-induced colitis in mice. High dose of TPs (HTPs) could prevent the colon from shortening, reduce activity of colonic myeloperoxidase and serum diamine oxidase (DAO), decrease the concentration of D-lactate, and alleviate the colonic tissue damage in colitic mice. HTPs treatment stimulated Foxp3+T cells, and promoted the production of anti-inflammatory cytokines whereas it reduced the production of pro-inflammatory and the portion of immunoglobulin A (IgA)-coated bacteria, which was related to modulation of immune responses. 16S rRNA sequencing analysis showed that TPs could significantly increase gut community diversity, and restore the relative abundances of Lactobacillus, Odoribacter, Helicobacter, Ruminococcaceae, and Marinifilaceae. According to metabolomic analysis, HTPs induced specific microbial metabolites akin to that in normal mice. Tyrosine biosynthesis, tryptophan metabolism, and bile acid metabolism were influenced in the HTPs group compared with those in the DSS group. HTPs could alleviate DSS-induced colitis by immunoregulation and restored the gut microbiota and microbial metabolites. The results indicated that HTPs have potential to be developed as a food supplement to ameliorate intestinal diseases.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Basidiomycota/chemistry , Colitis/chemically induced , Colitis/drug therapy , Dextran Sulfate/adverse effects , Forkhead Transcription Factors/metabolism , Fungal Polysaccharides/administration & dosage , Gastrointestinal Microbiome/drug effects , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/metabolism , Protective Agents/administration & dosage , T-Lymphocytes, Regulatory/immunology , Animals , Basidiomycota/genetics , Bile Acids and Salts/metabolism , Colitis/immunology , Colitis/microbiology , Disease Models, Animal , Female , Fungal Polysaccharides/chemistry , Mice , Mice, Inbred C57BL , Molecular Weight , RNA, Ribosomal, 16S/genetics , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , Treatment Outcome , Tryptophan/metabolism , Tyrosine/biosynthesis
18.
Mol Nutr Food Res ; 65(6): e2000635, 2021 03.
Article in English | MEDLINE | ID: mdl-33559319

ABSTRACT

SCOPE: The transgenerational impact of dietary fat remains unclear. Here, the role of maternal fat consumption as a modulator of gut microbial communities and infectious disease outcomes in their offspring is explored. METHODS AND RESULTS: C57BL/6 mice are fed isocaloric high-fat diets throughout breeding, gestation and lactation. Diets contained either milk fat (MF), olive oil (OO) or corn oil (CO), with or without fish oil. The pups born to maternally exposed mice are weaned on to chow and raised into adulthood. At 8 weeks, the offsprings are either euthanized for colonic 16S rRNA analysis or challenged with the enteric pathogen, Citrobacter rodentium. Maternal CO exposure resulted in unique clustering of bacterial communities in offspring compared with MF and OO. Diets rich in CO reduced survival in offspring challenged with C. rodentium. The addition of fish oil did not improve mortality caused by CO and worsened disease outcomes when combined with OO. Unlike the unsaturated diets, MF is protective with and without fish oil. CONCLUSIONS: Overall, these data reveal that maternal intake of fatty acids do have transgenerational impacts on their offspring's bacteriome and enteric infection risk. Based on this study, saturated fats should be included in maternal diets.


Subject(s)
Colitis/immunology , Colitis/microbiology , Diet, High-Fat/adverse effects , Dietary Fats/pharmacology , Gastrointestinal Microbiome/physiology , Animals , Corn Oil/chemistry , Corn Oil/pharmacology , Cytokines/metabolism , Dietary Fats/adverse effects , Enterobacteriaceae Infections/immunology , Fatty Acids, Volatile/metabolism , Female , Fish Oils/chemistry , Fish Oils/pharmacology , Male , Mice, Inbred C57BL , Olive Oil/chemistry , Olive Oil/pharmacology , Polysaccharides/chemistry , Polysaccharides/metabolism , Risk Factors
19.
Nutrients ; 14(1)2021 Dec 28.
Article in English | MEDLINE | ID: mdl-35010997

ABSTRACT

Coix seed is a functional food in the Chinese diet that possesses the ability to alleviate ulcerative colitis clinically. However, the underlying mechanisms remain ambiguous. In this study, we investigated the protective effect of the Coix seed diet on experimental colitis mice. The mice were randomly divided into four groups: control group, model group, Coix seed feed group, and positive control group. The maintenance feed of the mice was replaced with Coix seed feed 10 days before orally administering the mice 5% (w/v) dextran sulfate sodium drink. As a result, the Coix seed feed alleviated colitis symptoms, maintained the complete blood count at a normal level, reduced the pathological score, relieved inflammatory cytokine secretion, and alleviated oxidative stress. Network pharmacology analysis was used for further exploration of the targets of Coix seed feed. The results showed that T-cell regulation is one of the targets of Coix seed feed, and the analysis of the T-lymphocyte subset and innate immune cell distribution of the colon tissue supported the network pharmacology results. In conclusion, Coix seed, as a staple food, can alleviate experimental colitis, and the mechanism may be related to the immune regulation effect of Coix seeds.


Subject(s)
Coix , Colitis/therapy , Drugs, Chinese Herbal/pharmacology , Immunity, Innate/drug effects , Seeds , Animal Feed , Animals , Colitis/chemically induced , Colitis/immunology , Colon/immunology , Dextran Sulfate , Disease Models, Animal , Functional Food , Mice , Network Pharmacology , T-Lymphocyte Subsets/drug effects
20.
J Allergy Clin Immunol ; 147(1): 335-348.e11, 2021 01.
Article in English | MEDLINE | ID: mdl-32407834

ABSTRACT

BACKGROUND: The cross-talk between the host and its microbiota plays a key role in the promotion of health. The production of metabolites such as polyamines by intestinal-resident bacteria is part of this symbiosis shaping host immunity. The polyamines putrescine, spermine, and spermidine are abundant within the gastrointestinal tract and might substantially contribute to gut immunity. OBJECTIVE: We aimed to characterize the polyamine spermidine as a modulator of T-cell differentiation and function. METHODS: Naive T cells were isolated from wild-type mice or cord blood from healthy donors and submitted to polarizing cytokines, with and without spermidine treatment, to evaluate CD4+ T-cell differentiation in vitro. Moreover, mice were subjected to oral supplementation of spermidine, or its precursor l-arginine, to assess the frequency and total numbers of regulatory T (Treg) cells in vivo. RESULTS: Spermidine modulates CD4+ T-cell differentiation in vitro, preferentially committing naive T cells to a regulatory phenotype. After spermidine treatment, activated T cells lacking the autophagy gene Atg5 fail to upregulate Foxp3 to the same extent as wild-type cells. These results indicate that spermidine's polarizing effect requires an intact autophagic machinery. Furthermore, dietary supplementation with spermidine promotes homeostatic differentiation of Treg cells within the gut and reduces pathology in a model of T-cell transfer-induced colitis. CONCLUSION: Altogether, our results highlight the beneficial effects of spermidine, or l-arginine, on gut immunity by promoting Treg cell development.


Subject(s)
Cell Differentiation/drug effects , Colitis/immunology , Immunity, Mucosal/drug effects , Spermidine/pharmacology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL