Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 444
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Transpl Immunol ; 84: 102044, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663757

ABSTRACT

BACKGROUND: Glutamine is crucial for the activation and efficacy of T cells, and may play a role in regulating the immune environment. This study aimed to investigate the potential role of glutamine in the activation and proliferation of induced regulatory T cells (iTregs). METHODS: CD4+CD45RA+T cells were sorted from peripheral blood mononuclear cells and cultured to analyze iTreg differentiation. Glutamine was then added to the culture system to evaluate the effects of glutamine on iTregs by determining oxidative phosphorylation (OXPHOS), apoptosis, and cytokine secretion. Additionally, a humanized murine graft-versus-host disease (GVHD) model was constructed to confirm the efficacy of glutamine-treated iTregs in vivo. RESULTS: After being cultured in vitro, glutamine significantly enhanced the levels of Foxp3, CTLA-4, CD39, CD69, IL-10, TGF-ß, and Ki67 (CTLA-4, IL-10, TGF-ß are immunosuppressive markers of iTregs) compared with that of the control iTregs (P < 0.05). Furthermore, the growth curve showed that the proliferative ability of glutamine-treated iTregs was better than that of the control iTregs (P < 0.01). Compared with the control iTregs, glutamine supplementation significantly increased oxygen consumption rates and ATP production (P < 0.05), significantly downregulated Annexin V and Caspase 3, and upregulated BCL2 (P < 0.05). However, GPNA significantly reversed the effects of glutamine (P < 0.05). Finally, a xeno-GVHD mouse model was successfully established to confirm that glutamine-treated iTregs increased the mice survival rate, delayed weight loss, and alleviated colon injury. CONCLUSION: Glutamine supplementation can improve the activity and immunosuppressive action of iTregs, and the possible mechanisms by which this occurs are related to cell proliferation, apoptosis, and OXPHOS.


Subject(s)
Glutamine , Graft vs Host Disease , T-Lymphocytes, Regulatory , Glutamine/pharmacology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Animals , Mice , Humans , Cells, Cultured , Graft vs Host Disease/immunology , Cell Proliferation/drug effects , Lymphocyte Activation/drug effects , Disease Models, Animal , Apoptosis/drug effects , Cell Differentiation/drug effects , Immunosuppression Therapy , Cytokines/metabolism
2.
Chin J Nat Med ; 22(4): 341-355, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38658097

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disorder marked by persistent synovial inflammation and joint degradation, posing challenges in the development of effective treatments. Nuciferine, an alkaloid found in lotus leaf, has shown promising anti-inflammatory and anti-tumor effects, yet its efficacy in RA treatment remains unexplored. This study investigated the antiproliferative effects of nuciferine on the MH7A cell line, a human RA-derived fibroblast-like synoviocyte, revealing its ability to inhibit cell proliferation, promote apoptosis, induce apoptosis, and cause G1/S phase arrest. Additionally, nuciferine significantly reduced the migration and invasion capabilities of MH7A cells. The therapeutic potential of nuciferine was further evaluated in a collagen-induced arthritis (CIA) rat model, where it markedly alleviated joint swelling, synovial hyperplasia, cartilage injury, and inflammatory infiltration. Nuciferine also improved collagen-induced bone erosion, decreased pro-inflammatory cytokines and serum immunoglobulins (IgG, IgG1, IgG2a), and restored the balance between T helper (Th) 17 and regulatory T cells in the spleen of CIA rats. These results indicate that nuciferine may offer therapeutic advantages for RA by decreasing the proliferation and invasiveness of FLS cells and correcting the Th17/Treg cell imbalance in CIA rats.


Subject(s)
Aporphines , Cell Proliferation , Synoviocytes , T-Lymphocytes, Regulatory , Th17 Cells , Animals , Cell Proliferation/drug effects , Synoviocytes/drug effects , Rats , Humans , Th17 Cells/drug effects , Th17 Cells/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Aporphines/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Male , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Fibroblasts/drug effects , Collagen , Apoptosis/drug effects , Cell Line
3.
Phytomedicine ; 128: 155490, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38460358

ABSTRACT

BACKGROUND: Nauclea officinalis (Pierre ex Pit.) Merr. & Chun (Rubiaceae) is widely used to treat respiratory diseases in China. Strictosamide is its main active component and has significant anti-inflammatory activity. However, the effects and molecular mechanisms of strictosamide in the treatment of acute lung injury (ALI) remain largely unknown. PURPOSE: This study aimed to examine the regulatory effects of strictosamide on T helper 17 cells (Th17 cells)/Regulatory T cells (Treg cells) and gut microbiota in ALI-affected mice. MATERIALS AND METHODS: The ALI model was induced using lipopolysaccharide (LPS) intraperitoneal injection. Hematoxylin-eosin (H&E) staining, the number of inflammatory cells in broncho-alveolar lavage fluid (BALF), the Wet/Dry (W/D) ratio, and myeloperoxidase (MPO) activity were utilized as evaluation indices for the therapeutic efficacy of strictosamide on ALI. Flow cytometry (FCM), enzyme-linked immune sorbent assay (ELISA), quantitative reverse transcription polymerase chain reaction (qRT-PCR), and western blotting were used to determine the regulation of strictosamide on the Th17/Treg cells and the STAT3/STAT5 signaling pathway. The analysis of gut microbiota was conducted using 16S rDNA sequencing. The verification of the relationship between the gut microbiome and immune function was conducted using Spearman analysis. RESULTS: Strictosamide attenuated inflammation on ALI induced by LPS, which reduced the levels of Th17-related factors interleukin (IL)-6 and IL-17 and increased Treg-related factors IL-10 and transforming growth factor (TGF)-ß. In the spleens and whole blood, strictosamide reduced the proportion of Th17 cells and increased the proportion of Treg cells. Furthermore, strictosamide increased Forkhead/winged helix transcription factor 3 (Foxp3) and p-STAT5 protein expression while inhibiting Retinoid-related orphan nuclear receptors-γt (RORγt) and p-STAT3 expression. Moreover, strictosamide reshaped the diversity and structure of the gut microbiota, and influence the associations between immune parameters and gut microbiota in ALI mice. CONCLUSIONS: In summary, the results of the current investigation showed that strictosamide has a therapeutic impact on LPS-induced ALI. The mechanism of action of this effect may be associated with the modulation of Th17 and Treg cells differentiation via the SATA signaling pathway, as well as the impact of the gut microbiota.


Subject(s)
Acute Lung Injury , Gastrointestinal Microbiome , Lipopolysaccharides , STAT3 Transcription Factor , T-Lymphocytes, Regulatory , Th17 Cells , Animals , Acute Lung Injury/drug therapy , T-Lymphocytes, Regulatory/drug effects , Gastrointestinal Microbiome/drug effects , Th17 Cells/drug effects , Male , Mice , STAT3 Transcription Factor/metabolism , Disease Models, Animal , Mice, Inbred BALB C , Mice, Inbred C57BL , Anti-Inflammatory Agents/pharmacology , Bronchoalveolar Lavage Fluid/cytology
4.
Int J Mol Sci ; 23(5)2022 Mar 06.
Article in English | MEDLINE | ID: mdl-35270015

ABSTRACT

Almost two years have passed since the outbreak reported for the first time in Wuhan of coronavirus disease 2019 (COVID-19), due to severe acute respiratory syndrome (SARS)-CoV-2 coronavirus, rapidly evolved into a pandemic. This infectious disease has stressed global health care systems. The mortality rate is higher, particularly in elderly population and in patients with comorbidities such as hypertension, diabetes mellitus, cardiovascular disease, chronic lung disease, chronic renal disease, and malignancy. Among them, subjects with diabetes have a high risk of developing severe form of COVID-19 and show increased mortality. How diabetes contributes to COVID-19 severity remains unclear. It has been hypothesized that it may be correlated with the effects of hyperglycemia on systemic inflammatory responses and immune system dysfunction. Vitamin D (VD) is a modulator of immune-response. Data from literature showed that vitamin D deficiency in COVID-19 patients increases COVID-19 severity, likely because of its negative impact on immune and inflammatory responses. Therefore, the use of vitamin D might play a role in some aspects of the infection, particularly the inflammatory state and the immune system function of patients. Moreover, a piece of evidence highlighted a link among vitamin D deficiency, obesity and diabetes, all factors associated with COVID-19 severity. Given this background, we performed an overview of the systematic reviews to assess the association between vitamin D supplementation and inflammatory markers in patients with diabetes; furthermore, vitamin D's possible role in COVID-19 patients was assessed as well. Three databases, namely MEDLINE, PubMed Central and the Cochrane Library of Systematic Reviews, were reviewed to retrieve the pertinent data. The aim of this review is to provide insight into the recent advances about the molecular basis of the relationship between vitamin D, immune response, inflammation, diabetes and COVID-19.


Subject(s)
COVID-19/immunology , Diabetes Mellitus/immunology , Immune System/immunology , Inflammation/immunology , Obesity/immunology , Vitamin D/immunology , COVID-19/virology , Humans , Immune System/drug effects , Meta-Analysis as Topic , SARS-CoV-2/physiology , Systematic Reviews as Topic , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Vitamin D/administration & dosage , Vitamins/administration & dosage , Vitamins/immunology
5.
J Neuroimmunol ; 362: 577778, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34814011

ABSTRACT

D-mannose (D-m) is a glucose epimer found in natural products, especially fruits. In mouse models of diabetes and airway inflammation, D-m supplementation via drinking water attenuated pathology by modifying cellular energy metabolism, leading to the activation of latent transforming growth factor beta (TGF-ß), which in turn induced T regulatory cells (Tregs). Given that Tregs are important in controlling neuroinflammation in experimental autoimmune encephalomyelitis (EAE) and likely in multiple sclerosis (MS), we hypothesized that D-m could also suppress EAE. We found that D-m delayed disease onset and reduced disease severity in two models of EAE. Importantly, D-m treatment prevented relapses in a relapsing-remitting model of EAE, which mimics the most common clinical manifestation of MS. EAE suppression was accompanied by increased frequency of CD4+FoxP3+ Tregs in the central nervous system, suggesting that EAE suppression resulted from Treg cell induction by D-m. These findings suggest that D-m has the potential to be a safe and low-cost complementary therapy for MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mannose/pharmacology , T-Lymphocytes, Regulatory/drug effects , Administration, Oral , Animals , Female , Mice
6.
Int Arch Allergy Immunol ; 183(1): 1-13, 2022.
Article in English | MEDLINE | ID: mdl-34818253

ABSTRACT

BACKGROUND: Juzentaihoto (JTT) is a Kampo prescription that has been used clinically for treating skin diseases such as atopic dermatitis in Japan. We have previously studied the anti-allergic effects of JTT on 2,4,6-trinitrochlorobenzene (TNCB)-induced contact hypersensitivity (CHS) in mice and demonstrated that it significantly suppresses ear swelling in a dose-dependent manner. However, the mechanism underlying the anti-allergic actions of JTT is obscure. METHODS: We investigated the mechanism underlying the anti-allergic effects of JTT using a TNCB-induced murine CHS model and adoptive cell transfer experiments. RESULTS: We showed that the anti-allergic effects of JTT are due to inhibition of effector T-cell activation and induction and/or activation of regulatory T cells. Furthermore, ex vivo experiments confirmed the effect of JTT on the activation of effector T cells and regulatory T cells, as interferon-γ production decreased, whereas interleukin (IL)-10 production increased, in the cultured lymphocytes obtained from 5% TNCB-sensitized mice treated with anti-CD3ε and anti-CD28 monoclonal antibodies. Flow cytometry showed that the CD4+CD25+Foxp3+, CD4+CD25+Foxp3-, and CD8+CD122+ cell population increased after oral administration of JTT. Finally, the anti-allergic effect of JTT by inducing and/or activating regulatory T cells (Tregs) was confirmed to be mediated by IL-10 through in vivo neutralization experiments with anti-IL-10 monoclonal antibodies. CONCLUSION: We suggested that JTT exerts anti-allergic effects by regulating the activation of effector T cells and Tregs involved in murine CHS model.


Subject(s)
Anti-Allergic Agents/pharmacology , Dermatitis, Allergic Contact/etiology , Drugs, Chinese Herbal/pharmacology , Lymphocyte Activation/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Administration, Oral , Adoptive Transfer , Animals , Anti-Allergic Agents/administration & dosage , Anti-Allergic Agents/chemistry , Biomarkers , Cytokines , Dermatitis, Allergic Contact/drug therapy , Dermatitis, Allergic Contact/metabolism , Disease Management , Disease Models, Animal , Disease Susceptibility , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/chemistry , Female , Immunophenotyping , Japan , Mice , T-Lymphocytes, Regulatory/metabolism , Treatment Outcome
7.
Can Respir J ; 2021: 6406295, 2021.
Article in English | MEDLINE | ID: mdl-34630778

ABSTRACT

Aim: To investigate the therapeutic effect of LiuJunZi decoction (LJZD) in an experimental model of asthma and uncover its potential mechanism. Materials and Methods: The ovalbumin (OVA) was applied to induce asthma in Balb/C mice, and LJZD was orally administrated to asthmatic mice. The lung function and histological lesion were evaluated by airway hyperresponsiveness assay, lung edema assay, and hematoxylin and eosin staining. The amounts of CD4+CD25+Foxp3+ TReg cells were analyzed through combining fluorescent antibody staining with flow cytometry assay. The levels of inflammatory factors and immunoglobulins were detected by enzyme-linked immuno sorbent assay (ELISA). The expression of miR-21 and miR-146a was investigated by real-time PCR. The protein expression of activating protein-1 (AP-1), nuclear factor kappa-B (NF-κB), and NF-κB inhibitor alpha (IκBα) was determined by western blotting. Results: LJZD improves OVA-induced asthma in Balb/C mice, which is manifested by decreasing lung edema, Penh levels, lung histological lesion, and inflammatory cell infiltration. LJZD increased the number of CD4+CD25+Foxp3+ TReg cells in blood mononuclear cells from asthmatic mice. Furthermore, LJZD reduced the levels of tumor necrosis factor-α (TNF-α), interleukin- (IL-) 4, IL-6, IgG1, and IgE, but increased interferon gamma (IFN-γ) expression, in serum of asthmatic mice, and also decreased the expression of IL-17a, IL-23, IL-25, and thymic stromal lymphopoietin (Tslp) in lung tissues. In addition, miR-21 and miR-146a expression and phospho (p)-NF-κB, p-IκBα, and AP-1 protein expression were inhibited by LJZD in lung tissues from asthmatic mice. Conclusion: LJZD improved OVA-induced asthma in Balb/C mice by inhibiting allergic inflammation and Th2 immunoreaction, which might be associated with the inactivation of the NF-κB signaling pathway.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Drugs, Chinese Herbal/administration & dosage , Ovalbumin/adverse effects , T-Lymphocytes, Regulatory/drug effects , Th2 Cells/drug effects , Animals , Anti-Asthmatic Agents/pharmacology , Asthma/chemically induced , Asthma/immunology , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Mice , Mice, Inbred BALB C , MicroRNAs , Ovalbumin/administration & dosage , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology
8.
BMC Complement Med Ther ; 21(1): 220, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34479568

ABSTRACT

BACKGROUND: ECa 233 is a standardized extract of C. asiatica containing the triterpenoid glycosides, madecassoside to asiaticoside in the ratio of (1.5 ± 0.5):1. Anti-inflammatory activities of ECa 233 have been reported; however the immunomodulatory effects of ECa 233 on regulatory T cells, which have a pivotal role in immune regulation, has not been elucidated. Therefore, we investigated the effects of ECa 233 on regulatory T cells that may provide benefits in autoimmune and chronic inflammatory diseases. METHODS: ECa 233 was prepared as oral suspension in 0.5% carboxymethylcellulose and administered to male Wistar rats via oral gavage. The pharmacokinetics and toxicity of ECa 233 were evaluated. Splenic lymphocytes were isolated and analyzed by flow cytometry and qPCR to determine the immunomodulatory effects of ECa 233 on regulatory T cells. RESULTS: All rats had good tolerability to ECa 233 and other test preparations. The pharmacokinetic study showed low oral bioavailability for both triterpenoids, with the maximum plasma concentration reached at 4 h for asiaticoside and at 0.5 h for madecassoside. Multiple oral administration of ECa 233 reduced the frequency of T cells, particularly CD8 T cells in rats. ECa 233 enhanced the percentage of regulatory T cells, characterized by high expression of CD25+ and upregulation of FoxP3 gene. CONCLUSIONS: The present study demonstrated that ECa 233 possesses immunosuppressive properties by enhancing regulatory T cells. These results provide in vivo evidence for the anti-inflammatory action of ECa 233, in line with previously reports, and the potential uses of ECa 233 in the treatment of chronic inflammatory and autoimmune diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Immunomodulation/drug effects , Plant Extracts/pharmacology , T-Lymphocytes, Regulatory/drug effects , Triterpenes/pharmacology , Animals , Rats
9.
Cell Commun Signal ; 19(1): 89, 2021 08 30.
Article in English | MEDLINE | ID: mdl-34461944

ABSTRACT

BACKGROUND: Metastasis represents the leading cause of death in patients with breast cancer. Traditional Chinese medicine is particularly appreciated for metastatic diseases in Asian countries due to its benefits for survival period prolongation and immune balance modulation. However, the underlying molecular mechanisms remain largely unknown. This study aimed to explore the antimetastatic effect and immunomodulatory function of a clinical formula Aiduqing (ADQ). METHODS: Naive CD4+ T cells, regulatory T cells (Tregs), and CD8+ T cells were sorted by flow cytometry. Then, breast cancer cells and these immune cells were co-cultured in vitro or co-injected into mice in vivo to simulate their coexistence. Flow cytometry, ELISA, qPCR, double luciferase reporter gene assay, and chromatin immunoprecipitation assay were conducted to investigate the immunomodulatory and antimetastatic mechanisms of ADQ. RESULTS: ADQ treatment by oral gavage significantly suppressed 4T1-Luc xenograft growth and lung metastasis in the orthotopic breast cancer mouse model, without noticeable hepatotoxicity, nephrotoxicity, or hematotoxicity. Meanwhile, ADQ remodeled the immunosuppressive tumor microenvironment (TME) by increasing the infiltration of tumor-infiltrating lymphocytes (TILs) and cytotoxic CD8+ T cells, and decreasing the infiltration of Tregs, naive CD4+ T cells, and tumor-associated macrophages (TAMs). Molecular mechanism studies revealed that ADQ remarkably inhibited CXCL1 expression and secretion from TAMs and thus suppressed the chemotaxis and differentiation of naive CD4+ T cells into Tregs, leading to the enhanced cytotoxic effects of CD8+ T cells. Mechanistically, TAM-derived CXCL1 promoted the differentiation of naive CD4+ T cells into Tregs by transcriptionally activating the NF-κB/FOXP3 signaling. Lastly, mouse 4T1-Luc xenograft experiments validated that ADQ formula inhibited breast cancer immune escape and lung metastasis by suppressing the TAM/CXCL1/Treg pathway. CONCLUSIONS: This study not only provides preclinical evidence supporting the application of ADQ in inhibiting breast cancer metastasis but also sheds novel insights into TAM/CXCL1/NF-κB/FOXP3 signaling as a promising therapeutic target for Treg modulation and breast cancer immunotherapy. Video Abstract.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Chemokine CXCL1/genetics , Medicine, Chinese Traditional , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Mice , Neoplasm Metastasis , T-Lymphocytes, Regulatory/drug effects , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/drug effects , Xenograft Model Antitumor Assays
10.
Int J Mol Sci ; 22(13)2021 Jun 26.
Article in English | MEDLINE | ID: mdl-34206987

ABSTRACT

Hepatitis C virus (HCV) is one of the main triggers of chronic liver disease. Despite tremendous progress in the HCV field, there is still no vaccine against this virus. Potential vaccines can be based on its recombinant proteins. To increase the humoral and, especially, cellular immune response to them, more effective adjuvants are needed. Here, we evaluated a panel of compounds as potential adjuvants using the HCV NS5B protein as an immunogen. These compounds included inhibitors of polyamine biosynthesis and urea cycle, the mTOR pathway, antioxidants, and cellular receptors. A pronounced stimulation of cell proliferation and interferon-γ (IFN-γ) secretion in response to concanavalin A was shown for antioxidant N-acetylcysteine (NAC), polyamine biosynthesis inhibitor 2-difluoromethylornithine (DFMO), and TLR9 agonist CpG ODN 1826 (CpG). Their usage during the immunization of mice with the recombinant NS5B protein significantly increased antibody titers, enhanced lymphocyte proliferation and IFN-γ production. NAC and CpG decreased relative Treg numbers; CpG increased the number of myeloid-derived suppressor cells (MDSCs), whereas neither NAC nor DFMO affected MDSC counts. NAC and DFMO suppressed NO and interleukin 10 (IL-10) production by splenocytes, while DFMO increased the levels of IL-12. This is the first evidence of immunomodulatory activity of NAC and DFMO during prophylactic immunization against infectious diseases.


Subject(s)
Acetylcysteine/pharmacology , Adjuvants, Immunologic/pharmacology , Eflornithine/pharmacology , Hepatitis C/immunology , Immunity, Active/drug effects , Viral Nonstructural Proteins/immunology , Animals , Cell Proliferation , Cells, Cultured , Female , Immunogenicity, Vaccine/drug effects , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-12/metabolism , Mice , Mice, Inbred DBA , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Nitric Oxide/metabolism , Oligodeoxyribonucleotides/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Viral Hepatitis Vaccines/immunology
11.
Front Immunol ; 12: 623087, 2021.
Article in English | MEDLINE | ID: mdl-34262557

ABSTRACT

Background: Seasonal variations have been reported for immune markers. However, the relative contributions of sunlight and vitamin D variability on such seasonal changes are unknown. Objective: This double-blind, randomized, placebo-controlled trial tested whether daily 400 IU vitamin D3 supplementation affected short-term (12 weeks) and long-term (43 weeks) natural regulatory T cell (nTreg) populations in healthy participants. Design: 62 subjects were randomized equally to vitamin D versus placebo in March and assessed at baseline, April (4w), June (12w), September (25w) and January (43w). Circulating nTregs, ex vivo proliferation, IL-10 and IFN-γ productions were measured. Vitamin D metabolites and sunlight exposure were also assessed. Results: Mean serum 25-hydroxyvitamin D (25(OH)D) increased from 35.8(SD 3.0) to 65.3(2.6) nmol/L in April and remained above 75 nmol/L with vitamin D supplementation, whereas it increased from 36.4(3.2) to 49.8(3.5) nmol/L in June to fall back to 39.6(3.5) nmol/L in January with placebo. Immune markers varied similarly between groups according to the season, but independently of 25(OH)D. For nTregs, the mean (%CD3+CD4+CD127lo cells (SEM)) nadir observed in March (2.9(0.1)%) peaked in September at 4.0(0.2)%. Mean T cell proliferation peaked in June (33156(1813) CPM) returning to the nadir in January (17965(978) CPM), while IL-10 peaked in June and reached its nadir in September (median (IQR) of 262(283) to (121(194) pg/ml, respectively). Vitamin D attenuated the seasonal increase in IFN-γ by ~28% with mean ng/ml (SEM) for placebo vs vitamin D, respectively, for April 12.5(1.4) vs 10.0(1.2) (p=0.02); June 13.9(1.3) vs 10.2(1.7) (p=0.02) and January 7.4(1.1) vs 6.0(1.1) (p=0.04). Conclusions: Daily low dose Vitamin D intake did not affect the nTregs population. There were seasonal variation in nTregs, proliferative response and cytokines, suggesting that environmental changes influence immune response, but the mechanism seems independent of vitamin D status. Vitamin D attenuated the seasonal change in T cell-produced IFN-γ, suggesting a decrease in effector response which could be associated with inflammation. Clinical Trial Registration: https://www.isrctn.com, identifier (ISRCTN 73114576).


Subject(s)
Cell Proliferation/drug effects , Cholecalciferol/administration & dosage , Cholecalciferol/immunology , Interferon-gamma/analysis , Seasons , T-Lymphocytes, Regulatory/immunology , Adult , Cholecalciferol/blood , Cholecalciferol/pharmacology , Dietary Supplements , Double-Blind Method , Female , Humans , Inflammation/immunology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-10/analysis , Interleukin-10/immunology , Male , Middle Aged , Sunlight , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/physiology
12.
Biomed Pharmacother ; 141: 111931, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34328111

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic inflammatory intestinal disorder that is difficult to cure and characterized by periods of relapse. To face the challenges of limited treatment strategies and drawbacks of conventional medications, developing new and promising strategies as well as safe and effective drugs for treatment of IBD has become an urgent demand for clinics. The imbalance of Th17/Treg is a crucial event for the development of IBD, and studies have verified that correcting the imbalance of Th17/Treg is an effective strategy for preventing and treating IBD. Recently, a growing body of studies has indicated that phytochemicals derived from natural products are potent regulators of Th17/Treg, and exert preferable protective benefits against colonic inflammation. In this review, the great potential of anti-colitis agents derived from natural products through targeting Th17/Treg cells and their action mechanisms for the treatment or prevention of IBD in recent research is summarized, which may help further the development of new drugs for IBD treatment.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Inflammatory Bowel Diseases/immunology , Phytochemicals/pharmacology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/therapeutic use , Humans , Inflammatory Bowel Diseases/drug therapy , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Phytochemicals/isolation & purification , Phytochemicals/therapeutic use , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects
13.
ACS Appl Mater Interfaces ; 13(31): 36824-36838, 2021 Aug 11.
Article in English | MEDLINE | ID: mdl-34314148

ABSTRACT

Although immuno-oncotherapy in clinic has gained great success, the immunosuppressive tumor microenvironment (TME) existing in the "cold" tumor with insufficient and exhausted lymphocytes may result in a lower-than-expected therapeutic efficiency. Therefore, a properly designed synergistic strategy that can effectively turn the "cold" tumor to "hot" should be considered to improve the therapeutic effects of immuno-oncotherapy. Herein, TME-responsive penetrating nanogels (NGs) were developed, which can improve the delivery and penetration of the co-loaded resiquimod (R848) and green tea catechin (EGCG) in tumors by a nano-sized controlled releasing system of the soluble cyclodextrin-drug inclusion complex. Consequently, the NGs effectively promoted the maturation of dendritic cells, stimulated the cytotoxic T lymphocytes (CTLs), and decreased the PD-L1 expression in tumors. The combination of NGs with the OX40 agonist (αOX40) further synergistically enhanced the activation and infiltration of CTLs into the deep tumor and inhibited the suppression effects from the regulatory T cells (Tregs). As a result, an increased ratio of active CTLs to Tregs in tumors (20.66-fold) was achieved with a 91.56% tumor suppression effect, indicating a successful switch of "cold" tumors to "hot" for an immunologically beneficial TME with significantly improved anti-tumor immune therapeutics. This strategy could be tailored to other immuno-oncotherapeutic approaches to solve the urgent efficiency concerns of the checkpoint-based treatment in clinic.


Subject(s)
Antineoplastic Agents/therapeutic use , Catechin/therapeutic use , Drug Carriers/chemistry , Imidazoles/therapeutic use , Nanogels/chemistry , Neoplasms/drug therapy , 2-Hydroxypropyl-beta-cyclodextrin/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , B7-H1 Antigen/metabolism , Catechin/chemistry , Catechin/pharmacokinetics , Cell Line, Tumor , Dendritic Cells/drug effects , Drug Carriers/pharmacokinetics , Drug Liberation , Female , Hyaluronic Acid/analogs & derivatives , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Immunomodulation , Mice, Inbred C57BL , Neoplasms/metabolism , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Regulatory/drug effects , Tumor Microenvironment/drug effects
14.
Am J Physiol Cell Physiol ; 321(3): C569-C584, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34288720

ABSTRACT

Rheumatoid arthritis (RA) is a debilitating autoimmune disease of unknown cause, characterized by infiltration and accumulation of activated immune cells in the synovial joints where cartilage and bone destructions occur. Myeloid-derived suppressor cells (MDSCs) are of myeloid origin and are able to suppress T cell responses. Src homology 2 domain-containing inositol polyphosphate 5-phosphatase 1 (SHIP1) was shown to be involved in the regulation of MDSC differentiation. The purpose of the present study was to investigate the effect of inhibition of SHIP1 on the expansion of MDSCs in RA using a collagen-induced inflammatory arthritis (CIA) mouse model. In DBA/1 mice, treatment with a small molecule-specific SHIP1 inhibitor 3α-aminocholestane (3AC) induced a marked expansion of MDSCs in vivo. Both pretreatment with 3AC of DBA/1 mice prior to CIA induction and intervention with 3AC during CIA progression significantly reduced disease incidence and severity. Adoptive transfer of MDSCs isolated from 3AC-treated mice, but not naïve MDSCs from normal mice, into CIA mice significantly reduced disease incidence and severity, indicating that the 3AC-induced MDSCs were the cellular mediators of the observed amelioration of the disease. In conclusion, inhibition of SHIP1 expands MDSCs in vivo and attenuates development of CIA in mice. Small molecule-specific inhibition of SHIP1 may therefore offer therapeutic benefit to patients with RA and other autoimmune diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/drug therapy , Cholestanes/pharmacology , Myeloid-Derived Suppressor Cells/immunology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Cell Communication , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Gene Expression , Humans , Joint Capsule/drug effects , Joint Capsule/immunology , Joint Capsule/pathology , Mice , Mice, Inbred DBA , Mice, Knockout , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/transplantation , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/antagonists & inhibitors , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/immunology , Severity of Illness Index , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology
15.
Immunohorizons ; 5(6): 424-437, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112701

ABSTRACT

Graft-versus-host disease (GVHD) remains to be a significant cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). IL-2-inducible T cell kinase (ITK), a TEC cytoplasmic tyrosine kinase, has an essential role in T cell development and receptor signaling. The ITK/Bruton tyrosine kinase inhibitor ibrutinib has been shown to improve chronic GVHD symptoms; however, the effect of ITK selective inhibition on acute GVHD remains unclear. In this study, we evaluated the pharmacological effects of an ITK selective inhibitor (ITKsi) on acute GVHD using murine bone marrow transplantation models. First, we found that CD4+ T cell differentiation toward Th1, Th2, or Th17 was inhibited following ITKsi treatment in a dose-dependent manner while maintaining regulatory T cells in the presence of alloantigens both in vitro and in vivo. ITKsi preferentially inhibited inflammatory cytokine production and in vivo proliferation of alloreactive T cells. We then demonstrated that short-term exposure of donor graft cells to ITKsi significantly delayed the onset of GVHD-associated mortality without compromising the donor cell engraftment and the graft-versus-tumor effect, indicating the potential of ITK selective inhibition in the setting of clinical allogeneic HSCT. These findings suggest that ITK is a potential therapeutic target against GVHD, and the pharmacological ITK inhibitor may serve as a novel strategy for immune regulation after HSCT.


Subject(s)
Bone Marrow Transplantation/adverse effects , Graft vs Host Disease/prevention & control , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Transplantation Conditioning/methods , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Graft vs Host Disease/immunology , Humans , Mice , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
16.
Front Immunol ; 12: 634509, 2021.
Article in English | MEDLINE | ID: mdl-33953711

ABSTRACT

Tregitopes (T regulatory epitopes) are IgG-derived peptides with high affinity to major histocompatibility complex class II (MHCII), that are known to promote tolerance by activating T regulatory cell (Treg) activity. Here we characterized the effect of IgG Tregitopes in a well-established murine model of allergic asthma, demonstrating in vivo antigen-specific tolerance via adoptive transfer of Tregitope-and-allergen-activated Tregs. Asthma is a heterogeneous chronic inflammatory condition affecting the airways and impacting over 300 million individuals worldwide. Treatment is suppressive, and no current therapy addresses immune regulation in severely affected asthmatics. Although high dose intra-venous immunoglobulin (IVIg) is not commonly used in the asthma clinic setting, it has been shown to improve severe asthma in children and in adults. In our laboratory, we previously demonstrated that IVIg abrogates airway hyperresponsiveness (AHR) in a murine model of asthma and induces suppressive antigen-specific T-regulatory cells. We hypothesized that IgG-derived Tregitopes would modulate allergic airway disease by inducing highly suppressive antigen-specific Tregs capable of diminishing T effector cell responses and establishing antigen-specific tolerance. Using ovalbumin (OVA-) and ragweed-driven murine models of allergic airway disease, we characterized the immunoregulatory properties of Tregitopes and performed Treg adoptive transfer to OVA- and ragweed-allergic mice to test for allergen specificity. Treatment with Tregitopes attenuated allergen-induced airway hyperresponsiveness and lung inflammation. We demonstrated that Tregitopes induce highly suppressive allergen-specific Tregs. The tolerogenic action of IgG Tregitopes in our model is very similar to that of IVIg, so we foresee that IgG Tregitopes could potentially replace steroid-based treatment and can offer a synthetic alternative to IVIg in a range of inflammatory and allergic conditions.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Epitopes, T-Lymphocyte/drug effects , Immunoglobulin Fab Fragments/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Lung/drug effects , Lymphocyte Activation/drug effects , T-Lymphocytes, Regulatory/drug effects , Adoptive Transfer , Animals , Animals, Genetically Modified , Antigens, Plant , Asthma/immunology , Asthma/metabolism , Asthma/physiopathology , Bronchoconstriction/drug effects , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Humans , Inflammation Mediators/metabolism , Lung/immunology , Lung/metabolism , Lung/physiopathology , Mice, Inbred C57BL , Ovalbumin , Plant Extracts , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation
17.
J Ethnopharmacol ; 276: 114178, 2021 Aug 10.
Article in English | MEDLINE | ID: mdl-33945857

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Si-Miao-Yong-An decoction (SMYAD) is a renowned traditional Chinese medicinal formula. SMYAD was originally recorded in the "Shi Shi Mi Lu", which was edited by medical scientist Chen Shi'duo during the Qing Dynasty. SMYAD has been traditionally used to treat thromboangiitis obliterans. At present, it is mainly used in clinical applications and research of cardiovascular diseases. AIM OF THE STUDY: To explore the effects of SMYAD on the pathological changes of atherosclerosis (AS) and the differentiation of monocytes, macrophages, and regulatory T (Treg) cells in apolipoprotein E knockout (ApoE-/-) mice. MATERIALS AND METHODS: Eight C57BL/6J mice, which were fed with normal diet for 16 weeks, were used as control group. Forty ApoE-/- mice were randomly divided into model group, atorvastatin group, SMYAD low-dose (SMYAD-LD) group, SMYAD medium-dose (SMYAD-MD) group, and SMYAD high-dose (SMYAD-HD) group. ApoE-/- mice were fed with western diet (WD) for 8 weeks, and the drugs were continuously administered for 8 weeks. The levels of serum total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) were measured by the esterase method. Morphological changes of the aortic sinus in mice were observed by hematoxylin-eosin (HE) staining, the lipid infiltration of the aorta and aortic sinus were observed by oil red O staining, and the spleen index was calculated. The proportion of Ly6Chigh and Ly6Clow monocyte subsets, macrophages, and their M1 phenotype, as well as Treg cells in spleen were measured by flow cytometry. The expressions of cluster of differentiation 36 (CD36), scavenger receptor A1 (SRA1), lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), F4/80, and fork head frame protein 3 (FOXP3) in aortic sinus were assessed by immunohistochemical staining. The serum levels of oxidized low density lipoprotein (ox-LDL), interleukin-1ß (IL-1ß), IL-18, transforming growth factor-ß (TGF-ß), and IL-10 were measured by enzyme-linked immunosorbent assays (ELISA). RESULTS: Compared with the model group, the level of serum TC and LDL-C decreased in the SMYAD group, the pathological changes of aortic sinus decreased, and lipid infiltration of aorta and aortic sinus also decreased. These decreases were accompanied by a significant downregulation of CD36, SRA1, and LOX-1. Furthermore, the proportions of Ly6Chigh pro-inflammatory monocyte subsets, macrophages, and their M1 phenotypes in spleen decreased significantly, while the proportion of Treg cells increased. In addition, while the expression of F4/80 decreased, the expression of FOXP3 increased in the aorta sinus. The levels of serum pro-inflammatory factors IL-1ß and IL-18 decreased. CONCLUSIONS: SMYAD can improve the pathological changes associated with AS and can inhibit lipid deposition in ApoE-/- mice induced by WD diet. The likely mechanism is the inhibition of the differentiation and recruitment of monocytes and macrophages, the promotion of the differentiation and recruitment of Treg cells, as well as the reduction of the secretion of pro-inflammatory factors.


Subject(s)
Apolipoproteins E/genetics , Cell Differentiation/drug effects , Drugs, Chinese Herbal/pharmacology , Macrophages/drug effects , Monocytes/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Aorta/metabolism , Aorta/pathology , CD36 Antigens/metabolism , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Cholesterol/blood , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cytokines/blood , Drugs, Chinese Herbal/therapeutic use , Forkhead Transcription Factors/metabolism , Lipoproteins, LDL/blood , Male , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Receptors, G-Protein-Coupled/metabolism , Scavenger Receptors, Class E/metabolism , Spleen/drug effects , Spleen/metabolism , Triglycerides/blood
18.
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
19.
Int J Mol Sci ; 22(8)2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33924467

ABSTRACT

Kurarinone is a flavanone, extracted from Sophora flavescens Aiton, with multiple biological effects. Here, we determine the therapeutic potential of kurarinone and elucidate the interplay between kurarinone and the autoimmune disease rheumatoid arthritis (RA). Arthritis was recapitulated by induction of bovine collagen II (CII) in DBA/1 mice as a collagen-induced arthritis (CIA) model. After the establishment of the CIA, kurarinone was given orally from day 21 to 42 (100 mg/kg/day) followed by determination of the severity based on a symptom scoring scale and with histopathology. Levels of cytokines, anti-CII antibodies, and the proliferation and lineages of T cells from the draining lymph nodes were measured using ELISA and flow cytometry, respectively. The expressional changes, including STAT1, STAT3, Nrf2, KEAP-1, and heme oxygenase-1 (HO-1) changes in the paw tissues, were evaluated by Western blot assay. Oxidative stress featured with malondiadehyde (MDA) and hydrogen peroxide (H2O2) activities in paw tissues were also evaluated. Results showed that kurarinone treatment reduced arthritis severity of CIA mice, as well as their levels of proinflammatory cytokines, TNF-α, IL-6, IFN-γ, and IL-17A, in the serum and paw tissues. T cell proliferation was also reduced by kurarinone even under the stimulation of CII and anti-CD3 antibody. In addition, kurarinone reduced STAT1 and STAT3 phosphorylation and the proportions of Th1 and Th17 cells in lymph nodes. Moreover, kurarinone suppressed the production of MDA and H2O2. All while promoting enzymatic activities of key antioxidant enzymes, SOD and GSH-Px. In the paw tissues, upregulation of Nrf-2 and HO-1, and downregulation of KEAP-1 were observed. Overall, kurarinone showed an anti-inflammatory effect by inhibiting Th1 and Th17 cell differentiation and an antioxidant effect exerted in part through activating the Nrf-2/KEAP-1 pathway. These beneficial effects in CIA mice contributed to the amelioration of their arthritis, indicating that kurarinone might be an adjunct treatment option for rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Flavonoids/therapeutic use , Animals , Antioxidants/metabolism , Cattle , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chickens , Collagen Type II , Cytokines/blood , Cytokines/metabolism , Female , Flavonoids/pharmacology , Glutathione Peroxidase/metabolism , Hydrogen Peroxide/metabolism , Inflammation Mediators/metabolism , Joints/drug effects , Joints/pathology , Lymph Nodes/drug effects , Lymph Nodes/pathology , Malondialdehyde/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Superoxide Dismutase/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
20.
Int Immunopharmacol ; 97: 107683, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33915494

ABSTRACT

Juglone, mainly isolates from the green walnut husks of Juglans mandshurica, exhibits anti-cancer and anti-inflammaroty activities. But its protection on ulcerative colitis (UC) has never been explored. In this study, we first evaluated whether juglone ameliorated UC, and investigated its effects on gut microbiota and Th17/Treg balance in DSS-induced UC mice model. The model was established by administrating 2.7% DSS for seven days. Juglone was given daily by gavage for ten days, once a day. The disease activity index (DAI) decrease and pathological characteristics improvement demonstrated that the UC in mice was alleviated by juglone. Juglone treatment significantly inhibited the protein levels of IL-6, TNF-α and IL-1ß, improved the protein expression of IL-10. In addition, juglone altered microbial diversity and gut microbiota composition, including the enhancement of the ratio of Firmicutes to Bacteroidota and the abundance of Actinobacteriota, and decrease of the abundance of Verrucomicrobiota. Juglone treatment also inhibited the protein expressions of IL-6, STAT3 and RORγt, meanwhile improved the protein level of FOXP3. Furthermore, juglone inhibited Th17 development and increased Treg generation, beneficial to Th17/Treg balance. Together, we herein provided the first evidence to support that juglone, especially the high dose, possibly protected mice against UC by modulating gut microbiota and restoring Th17/Treg homeostasis.


Subject(s)
Colitis, Ulcerative/drug therapy , Gastrointestinal Microbiome/drug effects , Naphthoquinones/pharmacology , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Colitis, Ulcerative/microbiology , Colon/drug effects , Colon/immunology , Colon/microbiology , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Disease Models, Animal , Drug Evaluation, Preclinical , Gastrointestinal Microbiome/immunology , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Mice , Naphthoquinones/therapeutic use , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL