Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.756
Filter
1.
FASEB J ; 38(10): e23667, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38742812

ABSTRACT

Immunity imbalance of T helper 17 (Th17)/regulatory T (Treg) cells is involved in the pathogenesis of Crohn's disease (CD). Complanatuside A (CA), a flavonol glycoside, exerts anti-inflammatory activities and our study aimed to identify its effect on TNBS-induced colitis and the possible mechanisms. We found that CA alleviated the symptoms of colitis in TNBS mice, as demonstrated by prevented weight loss and colon length shortening, as well as decreased disease activity index scores, inflammatory scores, and levels of proinflammatory factors. Flow cytometry analysis showed that CA markedly reduced the percentage of Th17 cells while increasing the percentage of Treg cells in TNBS mice. Under Th17 cell polarizing conditions, CA inhibited the differentiation of Th17 cells while the Treg cell differentiation was elevated under Treg cell polarizing conditions. Furthermore, it was observed that JAK2 interacted with CA through six hydrogen bonds via molecular docking. The phosphorylation of JAK2/STAT3 was reduced by CA, which might be correlated with the protective effect of CA on colitis. In conclusion, CA reduced the imbalance of Th17/Treg cells by inhibiting the JAK2/STAT3 signaling pathway in TNBS-induced colitis, which may provide novel strategies for CD treatment.


Subject(s)
Colitis , Janus Kinase 2 , STAT3 Transcription Factor , Signal Transduction , T-Lymphocytes, Regulatory , Th17 Cells , Trinitrobenzenesulfonic Acid , Animals , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism , Janus Kinase 2/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , STAT3 Transcription Factor/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Mice , Signal Transduction/drug effects , Trinitrobenzenesulfonic Acid/toxicity , Male , Mice, Inbred BALB C , Cell Differentiation/drug effects
2.
Cancer Cell ; 42(5): 797-814.e15, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38744246

ABSTRACT

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


Subject(s)
Colitis , Immune Checkpoint Inhibitors , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/pharmacology , Humans , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Animals , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Interferon-gamma/metabolism , Female , Single-Cell Analysis , Mice
3.
Respir Res ; 25(1): 196, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715030

ABSTRACT

BACKGROUND: The treatment response to corticosteroids in patients with sarcoidosis is highly variable. CD4+ T cells are central in sarcoid pathogenesis and their phenotype in peripheral blood (PB) associates with disease course. We hypothesized that the phenotype of circulating T cells in patients with sarcoidosis may correlate with the response to prednisone treatment. Therefore, we aimed to correlate frequencies and phenotypes of circulating T cells at baseline with the pulmonary function response at 3 and 12 months during prednisone treatment in patients with pulmonary sarcoidosis. METHODS: We used multi-color flow cytometry to quantify activation marker expression on PB T cell populations in 22 treatment-naïve patients and 21 healthy controls (HCs). Pulmonary function tests at baseline, 3 and 12 months were used to measure treatment effect. RESULTS: Patients with sarcoidosis showed an absolute forced vital capacity (FVC) increase of 14.2% predicted (± 10.6, p < 0.0001) between baseline and 3 months. Good response to prednisone (defined as absolute FVC increase of ≥ 10% predicted) was observed in 12 patients. CD4+ memory T cells and regulatory T cells from patients with sarcoidosis displayed an aberrant phenotype at baseline, compared to HCs. Good responders at 3 months had significantly increased baseline proportions of PD-1+CD4+ memory T cells and PD-1+ regulatory T cells, compared to poor responders and HCs. Moreover, decreased fractions of CD25+ cells and increased fractions of PD-1+ cells within the CD4+ memory T cell population correlated with ≥ 10% FVC increase at 12 months. During treatment, the aberrantly activated phenotype of memory and regulatory T cells reversed. CONCLUSIONS: Increased proportions of circulating PD-1+CD4+ memory T cells and PD-1+ regulatory T cells and decreased proportions of CD25+CD4+ memory T cells associate with good FVC response to prednisone in pulmonary sarcoidosis, representing promising new blood biomarkers for prednisone efficacy. TRIAL REGISTRATION: NL44805.078.13.


Subject(s)
Prednisone , Programmed Cell Death 1 Receptor , Sarcoidosis, Pulmonary , T-Lymphocytes, Regulatory , Humans , Male , Sarcoidosis, Pulmonary/drug therapy , Sarcoidosis, Pulmonary/blood , Sarcoidosis, Pulmonary/immunology , Sarcoidosis, Pulmonary/diagnosis , Female , Middle Aged , Prednisone/therapeutic use , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Adult , Treatment Outcome , Memory T Cells/drug effects , Memory T Cells/immunology , Memory T Cells/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Glucocorticoids/therapeutic use , Vital Capacity/drug effects , Aged
4.
Sci Rep ; 14(1): 10396, 2024 05 06.
Article in English | MEDLINE | ID: mdl-38710724

ABSTRACT

Regulatory T cells (Tregs) is a subtype of CD4+ T cells that produce an inhibitory action against effector cells. In the present work we interrogated genomic datasets to explore the transcriptomic profile of breast tumors with high expression of Tregs. Only 0.5% of the total transcriptome correlated with the presence of Tregs and only four transcripts, BIRC6, MAP3K2, USP4 and SMG1, were commonly shared among the different breast cancer subtypes. The combination of these genes predicted favorable outcome, and better prognosis in patients treated with checkpoint inhibitors. Twelve up-regulated genes coded for proteins expressed at the cell membrane that included functions related to neutrophil activation and regulation of macrophages. A positive association between MSR1 and CD80 with macrophages in basal-like tumors and between OLR1, ABCA1, ITGAV, CLEC5A and CD80 and macrophages in HER2 positive tumors was observed. Expression of some of the identified genes correlated with favorable outcome and response to checkpoint inhibitors: MSR1, CD80, OLR1, ABCA1, TMEM245, and ATP13A3 predicted outcome to anti PD(L)1 therapies, and MSR1, CD80, OLR1, ANO6, ABCA1, TMEM245, and ATP13A3 to anti CTLA4 therapies, including a subgroup of melanoma treated patients. In this article we provide evidence of genes strongly associated with the presence of Tregs that modulates the response to check point inhibitors.


Subject(s)
Breast Neoplasms , T-Lymphocytes, Regulatory , Transcriptome , Humans , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/drug effects , Female , Breast Neoplasms/genetics , Breast Neoplasms/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Profiling , Prognosis
5.
Front Immunol ; 15: 1375340, 2024.
Article in English | MEDLINE | ID: mdl-38711519

ABSTRACT

Allergic asthma is a widely prevalent inflammatory condition affecting people across the globe. T cells and their secretory cytokines are central to the pathogenesis of allergic asthma. Here, we have evaluated the anti-inflammatory impact of dimethyl fumarate (DMF) in allergic asthma with more focus on determining its effect on T cell responses in allergic asthma. By utilizing the ovalbumin (OVA)-induced allergic asthma model, we observed that DMF administration reduced the allergic asthma symptoms and IgE levels in the OVA-induced mice model. Histopathological analysis showed that DMF treatment in an OVA-induced animal model eased the inflammation in the nasal and bronchial tissues, with a particular decrease in the infiltration of immune cells. Additionally, RT-qPCR analysis exhibited that treatment of DMF in an OVA-induced model reduced the expression of inflammatory cytokine (IL4, IL13, and IL17) while augmenting anti-inflammatory IL10 and Foxp3 (forkhead box protein 3). Mechanistically, we found that DMF increased the expression of Foxp3 by exacerbating the expression of nuclear factor E2-related factor 2 (Nrf2), and the in-vitro activation of Foxp3+ Tregs leads to an escalated expression of Nrf2. Notably, CD4-specific Nrf2 deletion intensified the allergic asthma symptoms and reduced the in-vitro iTreg differentiation. Meanwhile, DMF failed to exert protective effects on OVA-induced allergic asthma in CD4-specific Nrf2 knock-out mice. Overall, our study illustrates that DMF enhances Nrf2 signaling in T cells to assist the differentiation of Tregs, which could improve the anti-inflammatory immune response in allergic asthma.


Subject(s)
Asthma , Dimethyl Fumarate , Disease Models, Animal , NF-E2-Related Factor 2 , Signal Transduction , T-Lymphocytes, Regulatory , Animals , Dimethyl Fumarate/pharmacology , Dimethyl Fumarate/therapeutic use , NF-E2-Related Factor 2/metabolism , Asthma/drug therapy , Asthma/immunology , Asthma/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Mice , Signal Transduction/drug effects , Ovalbumin/immunology , Cytokines/metabolism , Mice, Inbred C57BL , Mice, Knockout , Female , Mice, Inbred BALB C
6.
J Interferon Cytokine Res ; 44(5): 208-220, 2024 May.
Article in English | MEDLINE | ID: mdl-38691831

ABSTRACT

Intestinal damage and secondary bacterial translocation are caused by the inflammatory response induced by sepsis. Tongfu Lifei (TLF) decoction has a protective effect on sepsis-related gastrointestinal function injury. However, the relation between gut microbiota, immune barrier, and sepsis under the treatment of TLF have not been well clarified yet. Here, rats were subjected to cecal ligation and puncture (CLP) to create a sepsis model. Subsequently, the TLF decoction was given to CLP rats by gavage, fecal microbiota transplantation (FMT), and antibiotic were used as positive control. TLF suppressed the inflammatory response and improved the pathological changes in the intestines of CLP rats. Besides, TLF promoted the balance of the percentage of the Th17 and Treg cells. Intestinal barrier function was also improved by TLF through enhancing ZO-1, and Occludin and Claudin 1 expression, preventing the secondary translocation of other gut microbiota. TLF dramatically boosted the gut microbiota's alpha- and beta-diversity in CLP rats. Moreover, it increased the relative abundance of anti-inflammatory gut microbiota and changed the progress of the glucose metabolism. In short, TLF regulated the gut microbiota to balance the ratio of Th17/Treg cells, reducing the inflammation in serum and intestinal mucosal injury in rats.


Subject(s)
Drugs, Chinese Herbal , Gastrointestinal Microbiome , Intestinal Mucosa , Sepsis , T-Lymphocytes, Regulatory , Th17 Cells , Animals , Gastrointestinal Microbiome/drug effects , Sepsis/immunology , Sepsis/drug therapy , Sepsis/complications , Th17 Cells/immunology , Th17 Cells/drug effects , Rats , Drugs, Chinese Herbal/pharmacology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/microbiology , Male , Rats, Sprague-Dawley
7.
Mol Immunol ; 170: 110-118, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38653076

ABSTRACT

Sepsis is a systemic inflammatory response syndrome caused by trauma or infection, which can lead to multiple organ dysfunction. In severe cases, sepsis can also progress to septic shock and even death. Effective treatments for sepsis are still under development. This study aimed to determine if targeting the PI3K/Akt signaling with CAL-101, a PI3K p110δ inhibitor, could alleviate lipopolysaccharide (LPS)-induced sepsis and contribute to immune tolerance. Our findings indicated that CAL-101 treatment improved survival rates and alleviated the progression of LPS-induced sepsis. Compared to antibiotics, CAL-101 not only restored the Th17/regulatory T cells (Treg) balance but also enhanced Treg cell function. Additionally, CAL-101 promoted type 2 macrophage (M2) polarization, inhibited TNF-α secretion, and increased IL-10 secretion. Moreover, CAL-101 treatment reduced pyroptosis in peritoneal macrophages by inhibiting caspase-1/gasdermin D (GSDMD) activation. This study provides a mechanistic basis for future clinical exploration of targeted therapeutics and immunomodulatory strategies in the treatment of sepsis.


Subject(s)
Class I Phosphatidylinositol 3-Kinases , Lipopolysaccharides , Mice, Inbred C57BL , Pyroptosis , Sepsis , T-Lymphocytes, Regulatory , Th17 Cells , Animals , Pyroptosis/drug effects , Sepsis/immunology , Sepsis/drug therapy , Lipopolysaccharides/pharmacology , Mice , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/immunology , Th17 Cells/drug effects , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/metabolism , Macrophages/immunology , Macrophages/drug effects , Macrophages/metabolism , Male , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Signal Transduction/drug effects
8.
Pharmacol Res ; 203: 107174, 2024 May.
Article in English | MEDLINE | ID: mdl-38580185

ABSTRACT

The emergence of immune checkpoint inhibitors (ICIs) has revolutionized the clinical treatment for tumor. However, the low response rate of ICIs remains the major obstacle for curing patients and effective approaches for patients with primary or secondary resistance to ICIs remain lacking. In this study, immune stimulating agent unmethylated CG-enriched (CpG) oligodeoxynucleotide (ODN) was locally injected into the tumor to trigger a robust immune response to eradicate cancer cells, while anti-CD25 antibody was applied to remove immunosuppressive regulatory T cells, which further enhanced the host immune activity to attack tumor systematically. The combination of CpG and anti-CD25 antibody obtained notable regression in mouse melanoma model. Furthermore, rechallenge of tumor cells in the xenograft model has resulted in smaller tumor volume, which demonstrated that the combinational treatment enhanced the activity of memory T cells. Remarkably, this combinational therapy presented significant efficacy on multiple types of tumors as well and was able to prevent relapse of tumor partially. Taken together, our combinational immunotherapy provides a new avenue to enhance the clinical outcomes of patients who are insensitive or resistant to ICIs treatments.


Subject(s)
Oligodeoxyribonucleotides , T-Lymphocytes, Regulatory , Animals , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Oligodeoxyribonucleotides/therapeutic use , Oligodeoxyribonucleotides/pharmacology , Mice , Mice, Inbred C57BL , Female , Humans , Cell Line, Tumor , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Interleukin-2 Receptor alpha Subunit/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/therapy , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/therapy , Vaccination , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use
9.
RMD Open ; 10(2)2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38580347

ABSTRACT

BACKGROUND: Systemic sclerosis (SSc) is a chronic autoimmune disease, with impaired immune response, increased fibrosis and endothelial dysfunction. Regulatory T cells (Tregs), which are essential to control inflammation, tissue repair and autoimmunity, have a decreased frequency and impaired function in SSc patients. Low-dose interleukin-2 (IL-2LD) can expand and activate Tregs and has, therefore, a therapeutic potential in SSc. OBJECTIVE: We aimed to assess the safety and biological efficacy of IL-2LD in patients with SSc. METHODS: As part of the TRANSREG open-label phase IIa basket trial in multiple autoimmune diseases, we studied nine patients with SSc without severe organ involvement. Patients received 1 million international units (MIU)/day of IL-2 for 5 days, followed by fortnightly injections for 6 months. Laboratory and clinical evaluations were performed between baseline and month 6. RESULTS: At day 8, the primary endpoint (Treg frequency) was reached with a 1.8±0.5-fold increase of Treg levels among CD4+ T lymphocytes (p=0.0015). There were no significant changes in effector T cells nor in B cells. IL-2LD was well tolerated, and no serious adverse events related to treatment occurred. There was a globally stable measurement in the modified Rodnan skin score and Valentini score at month 6. Disease activity and severity measures, the quality of life evaluated by EuroQL-5D-5L and pulmonary function test parameters remained stable during the study period. CONCLUSION: IL-2LD at a dosage of 1 MIU/day safely and selectively activates and expands Tregs. Clinical signs remain stable during the study period. This opens the door to properly powered phase II efficacy trials investigating IL-2LD therapeutic efficacy in SSc.


Subject(s)
Interleukin-2 , Scleroderma, Systemic , T-Lymphocytes, Regulatory , Humans , Autoimmune Diseases/drug therapy , Interleukin-2/adverse effects , Interleukin-2/therapeutic use , Quality of Life , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/immunology , T-Lymphocytes, Regulatory/drug effects
10.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 107-112, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38678615

ABSTRACT

Parkinson's disease (PD) is defined as a progressive neurodegenerative disease in middle-aged and elderly people. The therapeutic effect of ω-3 PUFAs in several neurodegenerative diseases has been well recognized. Nevertheless, whether nutrition supplementing ω-3 PUFAs exerts a neuroprotective role in PD remains elusive. Bioinformatics revealed 2D chemical structural formula of three components. Mice received indicated treatment with saline, MPTP or ω-3 PUFAs according to grouping. Behavioral function of mice was measured through motor tests such as rearing, akinesia, and rotarod tests. OFT test measured anxiety-like behaviors of mice. Western blotting and TUNEL staining measured dopaminergic fibers and neurons of mice. Western blotting measured inflammation and apoptosis-related protein levels in mouse tissue. FACS measured iTreg cell proportion in colon and brain tissues of mice. ω-3 PUFAs repaired MPTP-stimulated motor function damage in PD mice. ω-3 PUFAs mitigated MPTP-stimulated comorbid anxiety in PD mice. ω-3 PUFAs relieved MPTP-stimulated deficits of dopaminergic fibers and neurons in PD mice. ω-3 PUFAs repressed MPTP-stimulated inflammation and apoptosis pathway activation in PD mice. ω-3 PUFAs repaired MPTP-stimulated immune function damage in PD mice. ω-3 PUFAs exert a protective role in PD mice through alleviating motor function impairment and neuroinflammation by increasing intestinal inducible Treg cells, which may provide a new direction for seeking targeted therapy plans for PD in humans.


Subject(s)
Disease Models, Animal , Fatty Acids, Omega-3 , Mice, Inbred C57BL , Parkinson Disease , T-Lymphocytes, Regulatory , Animals , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Mice , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/pathology , Male , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Apoptosis/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Intestines/drug effects , Intestines/pathology , Behavior, Animal/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Inflammation/pathology , Inflammation/drug therapy , Inflammation/metabolism
11.
Front Immunol ; 15: 1326066, 2024.
Article in English | MEDLINE | ID: mdl-38665907

ABSTRACT

Introduction: Defective interleukin-2 (IL-2) production contributes to immune system imbalance in patients with systemic erythematosus lupus (SLE). Recent clinical studies suggested that low-dose IL-2 treatment is beneficial for SLE and the therapeutic effect is associated with regulatory T cell (Treg) expansion. Pharmacological calcineurin inhibition induces a reduction in the number of Tregs because they require stimulation of T cell receptor signaling and IL-2 for optimal proliferation. However, the activation of T cell receptor signaling is partially dispensable for the expansion of Tregs, but not for that of conventional T cells if IL-2 is present. Aim: We examined whether addition of IL-2 restores the Treg proportion even with concurrent use of a calcineurin inhibitor and if the follicular helper T cell (Tfh) proportion is reduced in an SLE-like murine chronic graft versus host disease model. Methods: Using a parent-into-F1 model, we investigated the effect of IL-2 plus tacrolimus on Treg and Tfh proportions and the therapeutic effect. Results: Treatment with a combination of IL-2 and tacrolimus significantly delayed the initiation of proteinuria and decreased the urinary protein concentration, whereas tacrolimus or IL-2 monotherapy did not significantly attenuate proteinuria. Phosphorylation of signal transducer and activator of transcription 3, a positive regulator of Tfh differentiation, was reduced by combination treatment, whereas phosphorylation of signal transducer and activator of transcription 5, a negative regulator, was not reduced. Conclusion: Addition of calcineurin inhibitors as adjunct agents may be beneficial for IL-2-based treatment of lupus nephritis.


Subject(s)
Interleukin-2 , Lupus Nephritis , T-Lymphocytes, Regulatory , Tacrolimus , Animals , Tacrolimus/therapeutic use , Tacrolimus/pharmacology , Lupus Nephritis/drug therapy , Lupus Nephritis/immunology , Mice , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Disease Models, Animal , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Drug Therapy, Combination , Female , T Follicular Helper Cells/immunology , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/pharmacology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/metabolism , Calcineurin Inhibitors/therapeutic use , Calcineurin Inhibitors/pharmacology , Bronchiolitis Obliterans Syndrome
12.
Molecules ; 29(8)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38675621

ABSTRACT

Allogeneic hematopoietic cell transplantation (allo-HCT) is a highly effective, well-established treatment for patients with various hematologic malignancies and non-malignant diseases. The therapeutic benefits of allo-HCT are mediated by alloreactive T cells in donor grafts. However, there is a significant risk of graft-versus-host disease (GvHD), in which the donor T cells recognize recipient cells as foreign and attack healthy organs in addition to malignancies. We previously demonstrated that targeting JAK1/JAK2, mediators of interferon-gamma receptor (IFNGR) and IL-6 receptor signaling, in donor T cells using baricitinib and ruxolitinib results in a significant reduction in GvHD after allo-HCT. Furthermore, we showed that balanced inhibition of JAK1/JAK2 while sparing JAK3 is important for the optimal prevention of GvHD. Thus, we have generated novel JAK1/JAK2 inhibitors, termed WU derivatives, by modifying baricitinib. Our results show that WU derivatives have the potential to mitigate GvHD by upregulating regulatory T cells and immune reconstitution while reducing the frequencies of antigen-presenting cells (APCs) and CD80 expression on these APCs in our preclinical mouse model of allo-HCT. In addition, WU derivatives effectively downregulated CXCR3 and T-bet in primary murine T cells. In summary, we have generated novel JAK inhibitors that could serve as alternatives to baricitinib or ruxolitinib.


Subject(s)
Disease Models, Animal , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Janus Kinase 2 , Pyrazoles , Transplantation, Homologous , Animals , Graft vs Host Disease/prevention & control , Graft vs Host Disease/drug therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Mice , Janus Kinase 2/metabolism , Janus Kinase 2/antagonists & inhibitors , Pyrazoles/pharmacology , Purines/pharmacology , Janus Kinase Inhibitors/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Sulfonamides/pharmacology , Azetidines/pharmacology , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism
13.
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
14.
J Nanobiotechnology ; 22(1): 201, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38659058

ABSTRACT

The utilization of extracellular vesicles (EV) in immunotherapy, aiming at suppressing peripheral immune cells responsible for inflammation, has demonstrated significant efficacy in treating various inflammatory diseases. However, the clinical application of EV has faced challenges due to their inadequate targeting ability. In addition, most of the circulating EV would be cleared by the liver, resulting in a short biological half-life after systemic administration. Inspired by the natural microvesicles (MV, as a subset of large size EV) are originated and shed from the plasma membrane, we developed the immunosuppressive MV-mimetic (MVM) from endotoxin tolerant dendritic cells (DC) by a straightforward and effective extrusion approach, in which DC surface proteins were inherited for providing the homing ability to the spleen, while αCD3 antibodies were conjugated to the MVM membranes for specific targeting of T cells. The engineered MVM carried a large number of bioactive cargos from the parental cells, which exhibited a remarkable ability to promote the induction of regulatory T cells (Treg) and polarization of anti-inflammatory M2 macrophages. Mechanistically, the elevated Treg level by MVM was mediated due to the upregulation of miR-155-3p. Furthermore, it was observed that systemic and local immunosuppression was induced by MVM in models of sepsis and rheumatoid arthritis through the improvement of Treg and M2 macrophages. These findings reveal a promising cell-free strategy for managing inflammatory responses to infections or tissue injury, thereby maintaining immune homeostasis.


Subject(s)
Cell-Derived Microparticles , Dendritic Cells , Inflammation , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Mice , Inflammation/drug therapy , Cell-Derived Microparticles/metabolism , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Extracellular Vesicles , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Sepsis/immunology , Sepsis/drug therapy , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Humans , Immunotherapy/methods
15.
Chemosphere ; 357: 141957, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641296

ABSTRACT

The current study aimed to explore whether bisphenol A (BPA) exposure aggravated the decrease in Tregs induced by ovalbumin (OVA) in adolescent female mouse models of asthma, and whether the process was associated with mTOR-mediated signaling pathways and DNA methylation levels. A total of 40 female C57BL/6 mice at the age of four weeks were used and divided into five groups after 1 week of domestication. Each group consisted of eight mice: the control group, OVA group, OVA + BPA (0.1 µg mL-1) group, OVA + BPA (0.2 µg mL-1) group, and OVA + BPA (0.4 µg mL-1) group. Results revealed that Foxp3 protein levels decreased in the spleens of mice exposed to BPA compared to those in the OVA group. After an elevation in BPA dose, the mRNAs of methyltransferases (Dnmt1, Dnmt3a, and Dnmt3b) were gradually upregulated. The mechanism was related to the activity of TLR4/NF-κB and PI3K/Akt/mTOR signaling pathways and the enhancement of Foxp3 DNA methylation. Our results, collectively, provided a new view for studying the mechanisms underlying BPA exposure-induced immune dysfunction. Investigation of the regulatory mechanisms of DNA methylation in the abnormal Th immune response caused by BPA exposure could help reveal the causes and molecular mechanisms underlying the high incidence of allergic diseases in children in recent years.


Subject(s)
Benzhydryl Compounds , DNA Methylation , Forkhead Transcription Factors , Mice, Inbred C57BL , Phenols , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Spleen , T-Lymphocytes, Regulatory , TOR Serine-Threonine Kinases , Animals , Phenols/toxicity , Benzhydryl Compounds/toxicity , DNA Methylation/drug effects , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Mice , TOR Serine-Threonine Kinases/metabolism , Female , Spleen/drug effects , Spleen/metabolism , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Up-Regulation/drug effects , Asthma/chemically induced , Ovalbumin
16.
Ecotoxicol Environ Saf ; 277: 116350, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38653026

ABSTRACT

Inorganic arsenic is a well-established environmental toxicant linked to acute liver injury, fibrosis, and cancer. While oxidative stress, pyroptosis, and ferroptosis are known contributors, the role of PTEN-induced kinase 1 (PINK1)-mediated mitophagy in arsenic-induced hepatic immunotoxicity remains underexplored. Our study revealed that acute arsenic exposure prompts differentiation of hepatic dendritic cells (DCs) and T helper (Th) 1, Th2, Th17, and regulatory T (Treg) cells, alongside increased transcription factors and cytokines. Inorganic arsenic triggered liver redox imbalance, leading to elevated alanine transaminase (ALT), hydrogen peroxide (H2O2), malondialdehyde (MDA), and activation of nuclear factor erythroid 2-related factor (Nrf2)/heme oxygenase-1 (HO-1) pathway. PINK1-mediated mitophagy was initiated, and its inhibition exacerbates H2O2 accumulation while promoting DCs/Th1/Th2/Treg differentiation in the liver of arsenic-exposed mice. Mitoquinone (MitoQ) pretreatment relieved arsenic-induced acute liver injury and immune imbalance by activating Nrf2/HO-1 and PINK1-mediated mitophagy. To our knowledge, this is the first report identifying PINK1-mediated mitophagy as a protective factor against inorganic arsenic-induced hepatic DCs/Th1/Th2 differentiation. This study has provided new insights on the immunotoxicity of inorganic arsenic and established a foundation for exploring preventive and therapeutic strategies targeting PINK1-mediated mitophagy in acute liver injury. Consequently, the application of mitochondrial antioxidant MitoQ may offer a promising treatment for the metalloid-induced acute liver injury.


Subject(s)
Antioxidants , Arsenic , Cell Differentiation , Liver , Mitophagy , Organophosphorus Compounds , Protein Kinases , Animals , Mitophagy/drug effects , Mice , Liver/drug effects , Antioxidants/pharmacology , Antioxidants/metabolism , Protein Kinases/metabolism , Cell Differentiation/drug effects , Organophosphorus Compounds/toxicity , Organophosphorus Compounds/pharmacology , Arsenic/toxicity , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , Dendritic Cells/drug effects , Mice, Inbred C57BL , Mitochondria/drug effects , Male , T-Lymphocytes, Regulatory/drug effects , Chemical and Drug Induced Liver Injury/prevention & control , Oxidative Stress/drug effects
17.
Ecotoxicol Environ Saf ; 277: 116364, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38657461

ABSTRACT

The purpose of this study was to investigate the effect of Treg/Th1 imbalance in cadmium-induced lung injury and the potential protective effect of astilbin against cadmium-induced lung injury in chicken. Cadmium exposure significantly decreased T-AOC and GSH-Px levels and SOD activity in the chicken lung tissues. In contrast, it significantly increased the MDA and NO levels. These results indicate that cadmium triggers oxidative stress in lungs. Histopathological analysis revealed that cadmium exposure further induced infiltration of lymphocytes in the chicken lungs, indicating that cadmium causes pulmonary damage. Further analysis revealed that cadmium decreased the expression of IL-4 and IL-10 but increased those of IL-17, Foxp3, TNF-α, and TGF-ß, indicating that the exposure of cadmium induced the imbalance of Treg/Th1. Moreover, cadmium adversely affected chicken lung function by activating the NF-kB pathway and inducing expression of genes downstream to these pathways (COX-2, iNOS), associated with inflammatory injury in the lung tissue. Astilbin reduced cadmium-induced oxidative stress and inflammation in the lungs by increasing antioxidant enzyme activities and restoring Treg/Th1 balance. In conclusion, our results suggest that astilbin treatment alleviated the effects of cadmium-mediated lung injury in chickens by restoring the Treg/Th1 balance.


Subject(s)
Cadmium , Chickens , Flavonols , Lung Injury , Lung , Oxidative Stress , Signal Transduction , T-Lymphocytes, Regulatory , Animals , Cadmium/toxicity , Oxidative Stress/drug effects , Lung/drug effects , Lung/pathology , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , Flavonols/pharmacology , Lung Injury/chemically induced , Lung Injury/drug therapy
18.
J Cancer Res Ther ; 20(2): 718-725, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38687945

ABSTRACT

PURPOSE: The complex strategy of hypo-fractionated radiotherapy (HFRT) in combination with an immune checkpoint inhibitor (ICI) can stimulate a potential systemic antitumor response; however, the abscopal effect is always precluded by the tumor microenvironment, which may limit sufficient T-cell infiltration of distant nonirradiated tumors for certain kinds of inhibitory factors, such as regulatory T-cells (Tregs). Additionally, low-dose cyclophosphamide (LD-CYC) can specifically kill regulatory Tregs and strongly synergize antigen-specific immune responses, which could promote an abscopal effect. MATERIALS AND METHODS: We explored whether a triple regimen consisting of HFRT, ICI, and LD-CYC could achieve a better systemic antitumor response in bilateral mouse tumor models. RESULT: Our data demonstrate that LD-CYC combined with HFRT and antiprogrammed cell death ligand 1 (PDL-1) therapy could enhance the abscopal effect than only HFRT/antiPDL-1 or HFRT alone. Surprisingly, repeat CYC doses cannot further restrain tumor proliferation but can prolong murine overall survival, as revealed by the major pathologic responses. These results are associated with increased CD8 + effector T-cell infiltration, although LD-CYC did not upregulate PDL-1 expression in the tumor. CONCLUSIONS: Compared with traditional strategies, for the first time, we demonstrated that a triple treatment strategy remarkably increased the number of radiation-induced tumor-infiltrating CD8 + T-cells, effectively decreasing infiltrating Tregs, and promoting an abscopal effect. Thus, we describe a novel and effective therapeutic approach by combining multiple strategies to target several tumor-mediated immune inhibitory mechanisms.


Subject(s)
Cyclophosphamide , Immune Checkpoint Inhibitors , T-Lymphocytes, Regulatory , Tumor Microenvironment , Animals , Cyclophosphamide/pharmacology , Cyclophosphamide/administration & dosage , Cyclophosphamide/therapeutic use , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects , Tumor Microenvironment/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/radiation effects , Female , Combined Modality Therapy , Disease Models, Animal , Melanoma, Experimental/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/radiotherapy , Radiation, Ionizing , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Agents, Alkylating/administration & dosage , Mice, Inbred C57BL , Humans , Cell Line, Tumor
19.
Int Immunopharmacol ; 132: 112019, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38599099

ABSTRACT

OBJECTIVE: The current study aimed to assess the modulating effect of IL-2 encapsulated chitosan-nanoparticles (CSNPs) on the function of Treg cells through induction of type 1 diabetes (T1D). Treg cell function was monitored by the forkhead box P3 (FoxP3) and transforming growth factor beta (TGFß) levels, correlating them with blood glucose and serum insulin levels. MATERIALS AND METHODS: In this case-control study, a low dose of IL-2 (free and chitosan-loaded) was injected into a diabetic mice group. The levels of FoxP3 and TGF-ß 1 were assessed using Enzyme-Linked Immunosorbent Assay. In addition, blood glucose and serum insulin levels were determined. RESULTS: The mean glucose level decreased significantly after free rIL-2 or rIL-2 / CSNPs treatment. Meanwhile, the mean serum insulin level was significantly increased after treatment with free rIL-2 or rIL-2/CSNPs. The mean levels of FoxP3 and TGFß 1 were significantly increased with either free rIL-2 or rIL-2/CSNPs compared to the T1D untreated group (P < 0.001). In the treated mice group receiving free CSNPs, there was a significant negative correlation between glucose and insulin levels. Moreover, FoxP3 & TGFß 1 levels had a significant positive correlation. In treated mice groups with free rIL-2 and IL-2 CSNPs, there was a significant positive correlation between FoxP3 and glucose levels. A significant negative correlation was found after conducting a correlation between insulin level and FoxP3 in the T1D/ rIL-2 / CSNPs group. CONCLUSIONS: Low-dose IL-2 selectively modulates FoxP3 + Tregs, and TGFß 1 increases their levels. These results demonstrated that IL-2-free and chitosan-loaded nanoparticles can be therapeutic agents in T1D.


Subject(s)
Blood Glucose , Chitosan , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Forkhead Transcription Factors , Insulin , Interleukin-2 , Nanoparticles , T-Lymphocytes, Regulatory , Animals , Chitosan/chemistry , Chitosan/administration & dosage , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Interleukin-2/metabolism , Interleukin-2/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/immunology , Blood Glucose/drug effects , Mice , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/blood , Forkhead Transcription Factors/metabolism , Insulin/blood , Male , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/blood , Streptozocin , Humans
20.
Int Immunopharmacol ; 132: 111937, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38569427

ABSTRACT

Tuberculosis (TB) treatment requires a long therapeutic duration and induces adverse effects such as hepatotoxicity, causing discontinuation of treatment. Reduced adherence to TB medications elevates the risk of recurrence and the development of drug resistance. Additionally, severe cavitary TB with a high burden of Mycobacterium tuberculosis (Mtb) and inflammation-mediated tissue damage may need an extended treatment duration, resulting in a higher tendency of drug-induced toxicity. We previously reported that the administration of Lactobacillus sakei CVL-001 (L. sakei CVL-001) regulates inflammation and improves mucosal barrier function in a murine colitis model. Since accumulating evidence has reported the functional roles of probiotics in drug-induced liver injury and pulmonary inflammation, we employed a parabiotic form of the L. sakei CVL-001 to investigate whether this supplement may provide beneficial effects on the reduction in drug-induced liver damage and pulmonary inflammation during chemotherapy. Intriguingly, L. sakei CVL-001 administration slightly reduced Mtb burden without affecting lung inflammation and weight loss in both Mtb-resistant and -susceptible mice. Moreover, L. sakei CVL-001 decreased T cell-mediated inflammatory responses and increased regulatory T cells along with an elevated antigen-specific IL-10 production, suggesting that this parabiotic may restrain excessive inflammation during antibiotic treatment. Furthermore, the parabiotic intervention significantly reduced levels of alanine aminotransferase, an indicator of hepatotoxicity, and cell death in liver tissues. Collectively, our data suggest that L. sakei CVL-001 administration has the potential to be an adjunctive therapy by reducing pulmonary inflammation and liver damage during anti-TB drug treatment and may benefit adherence to TB medication in lengthy treatment.


Subject(s)
Latilactobacillus sakei , Mycobacterium tuberculosis , Probiotics , Animals , Probiotics/therapeutic use , Probiotics/administration & dosage , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/immunology , Mice , Pneumonia/drug therapy , Pneumonia/immunology , Antitubercular Agents/therapeutic use , Antitubercular Agents/adverse effects , Female , Tuberculosis/drug therapy , Tuberculosis/immunology , Mice, Inbred C57BL , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/etiology , Humans , Lung/pathology , Lung/drug effects , Lung/immunology , Lung/microbiology , Interleukin-10/metabolism , Mice, Inbred BALB C , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Liver/drug effects , Liver/pathology , Liver/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...