Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Biomed Sci ; 31(1): 33, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38532423

ABSTRACT

BACKGROUND: T cell receptor (TCR) signaling and T cell activation are tightly regulated by gatekeepers to maintain immune tolerance and avoid autoimmunity. The TRAIL receptor (TRAIL-R) is a TNF-family death receptor that transduces apoptotic signals to induce cell death. Recent studies have indicated that TRAIL-R regulates T cell-mediated immune responses by directly inhibiting T cell activation without inducing apoptosis; however, the distinct signaling pathway that regulates T cell activation remains unclear. In this study, we screened for intracellular TRAIL-R-binding proteins within T cells to explore the novel signaling pathway transduced by TRAIL-R that directly inhibits T cell activation. METHODS: Whole-transcriptome RNA sequencing was used to identify gene expression signatures associated with TRAIL-R signaling during T cell activation. High-throughput screening with mass spectrometry was used to identify the novel TRAIL-R binding proteins within T cells. Co-immunoprecipitation, lipid raft isolation, and confocal microscopic analyses were conducted to verify the association between TRAIL-R and the identified binding proteins within T cells. RESULTS: TRAIL engagement downregulated gene signatures in TCR signaling pathways and profoundly suppressed phosphorylation of TCR proximal tyrosine kinases without inducing cell death. The tyrosine phosphatase SHP-1 was identified as the major TRAIL-R binding protein within T cells, using high throughput mass spectrometry-based proteomics analysis. Furthermore, Lck was co-immunoprecipitated with the TRAIL-R/SHP-1 complex in the activated T cells. TRAIL engagement profoundly inhibited phosphorylation of Lck (Y394) and suppressed the recruitment of Lck into lipid rafts in the activated T cells, leading to the interruption of proximal TCR signaling and subsequent T cell activation. CONCLUSIONS: TRAIL-R associates with phosphatase SHP-1 and transduces a unique and distinct immune gatekeeper signal to repress TCR signaling and T cell activation via inactivating Lck. Thus, our results define TRAIL-R as a new class of immune checkpoint receptors for restraining T cell activation, and TRAIL-R/SHP-1 axis can serve as a potential therapeutic target for immune-mediated diseases.


Subject(s)
Receptors, Antigen, T-Cell , Receptors, TNF-Related Apoptosis-Inducing Ligand , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Antigen, T-Cell/metabolism , Jurkat Cells , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Signal Transduction , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Phosphorylation , Lymphocyte Activation , Tyrosine/metabolism
2.
J Immunol ; 208(7): 1534-1544, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35264458

ABSTRACT

Recent evidence from several autoimmune animal models has demonstrated that TRAIL suppresses the activation of T cells and inhibits autoimmune inflammation via an apoptosis-independent pathway. However, it remains unclear whether the immunosuppressive effects of TRAIL are dependent on its direct effects on T cells or on other immune cells to regulate T cells for the induction of disease. Therefore, we generated mice with T cell-specific TRAIL receptor (TRAIL-R) conditional knockout to investigate the impact of TRAIL on autoimmune inflammation and disease induction in experimental autoimmune encephalomyelitis (EAE). T cell-specific TRAIL-R knockout mice were found to completely reverse the TRAIL-mediated suppression of inflammation and disease induction, indicating that TRAIL-R on T cells is essential for TRAIL-mediated suppression of inflammation and disease induction in EAE. Moreover, the immune suppression effects were not due to the induction of cell apoptosis, but to the direct inhibition of T cell activation. In addition, RNA sequencing and transcriptome analysis revealed that TRAIL-R signaling significantly downregulated the genes involved in TCR signaling pathways, T cell differentiation, and proinflammatory cytokines. These results indicate that TRAIL-R on T cells is critical for pathologic T cell activation and induction of inflammation in EAE, suggesting that TRAIL-R serves as a novel immune checkpoint receptor in T cell-mediated autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Animals , Lymphocyte Activation , Mice , Mice, Inbred C57BL , T-Lymphocytes , TNF-Related Apoptosis-Inducing Ligand
3.
Biochem Pharmacol ; 193: 114760, 2021 11.
Article in English | MEDLINE | ID: mdl-34492272

ABSTRACT

The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway is characterized by diverse immune regulatory systems involving cell proliferation, survival, and inflammation and immune tolerance. Aberrant JAK/STAT transduction activates proinflammatory cytokine signaling that jeopardize the immune balance and thus contributes to the development of autoimmune diseases and cancer progression. The success of several small-molecule JAK inhibitors in the treatment of rheumatologic diseases demonstrates that targeting the JAK/STAT pathway is efficient in suppressing inflammation and sheds light on their therapeutic potential in several autoimmune diseases and cancers. In this review, we discuss the signal transduction and molecular mechanism involving immune function through the JAK-STAT pathway, outline the role of this pathway in autoimmunity and oncoimmunology, and explain the preclinical and clinical trial evidence for the therapeutic potential of targeting the JAK-STAT signaling pathway. Issues regarding the safety and clinical efficacy of JAK inhibitors are reviewed. Ongoing studies are addressed with a focus on emerging indications for JAK inhibition and explanations of the novel mechanisms of JAK-STAT signaling blockade.


Subject(s)
Autoimmune Diseases/metabolism , Janus Kinases/metabolism , Neoplasms/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/physiology , Animals , Autoimmune Diseases/genetics , Gene Expression Regulation , Humans , Janus Kinases/genetics , Neoplasms/genetics , STAT Transcription Factors/genetics
4.
Cancers (Basel) ; 13(6)2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33801815

ABSTRACT

Immune checkpoints play critical roles in the regulation of T-cell effector function, and the effectiveness of their inhibitors in cancer therapy has been established. Immune checkpoint inhibitors (ICIs) constitute a paradigm shift in cancer therapy in general and cancer immunotherapy in particular. Immunotherapy has been indicated to reinvigorate antitumor T-cell activity and dynamically modulate anticancer immune responses. However, despite the promising results in the use of immunotherapy in some cancers, numerous patients do not respond to ICIs without the existence of a clear predictive biomarker. Overall, immunotherapy involves a certain degree of uncertainty and complexity. Research on the exploration of cellular and molecular factors within the tumor microenvironment (TME) aims to identify possible mechanisms of immunotherapy resistance, as well as to develop novel combination strategies involving the specific targeting of the TME for cancer immunotherapy. The combination of this approach with other types of treatment, including immune checkpoint blockade therapy involving multiple agents, most of the responses and effects in cancer therapy could be significantly enhanced, but the appropriate combinations have yet to be established. Moreover, the in-depth exploration of complexity within the TME allows for the exploration of pathways of immune dysfunction. It may also aid in the identification of new therapeutic targets. This paper reviews recent advances in the improvement of therapeutic efficacy on the immune context of the TME and highlights its contribution to cancer immunotherapy.

5.
Cancers (Basel) ; 13(6)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33809137

ABSTRACT

Autophagy plays a crucial role in maintenance of cellular homeostasis via intracellular signaling pathways, lysosomal degradation of selective cargo and mediating protein secretion. Dysregulation of autophagy has been implicated in tumorigenesis, tumor progression, and resistance to therapy. However, the mechanism of autophagy-dependent secretion involved in the responsiveness to chemotherapy is poorly understood. In this study, we showed that mitoxantrone (MitoX), a chemotherapeutic agent used for treating various cancers but not melanoma, induced autophagy in melanoma cells in vitro and in vivo. We also found that plasminogen activator inhibitor (PAI)-1 secretion by MitoX-induced autophagy modulated the pro-tumoral microenvironment. Attenuation of PAI-1 activity using a specific inhibitor, tiplaxtinin (TPX), or by targeting the autophagy gene, Becn1, induced efficient antitumor immunity, thereby overcoming the resistance to MitoX in vivo. Of note, the therapeutic efficacy of TPX was abolished in MitoX-treated Becn1-defective tumors. Collectively, our results demonstrate that tumor autophagy-dependent PAI-1 secretion impairs the therapeutic efficacy of MitoX and highlight targeting of tumor autophagy or its secretory cargo, PAI-1, as a novel strategy to repurpose MitoX-based chemotherapy for melanoma treatment.

6.
J Formos Med Assoc ; 120(9): 1667-1675, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33836940

ABSTRACT

Systemic lupus erythematosus (SLE) is a complex, heterogeneous, systemic autoimmune disease involving a wide array of aberrant innate and adaptive immune responses. The immune microenvironment of SLE promotes the metabolic reprogramming of immune cells, leading to immune dyshomeostasis and triggering autoimmune inflammation. Different immune subsets switch from a resting state to a highly metabolic active state by alternating the redox-sensitive signaling pathway and the involved metabolic intermediates to amplify the inflammatory response, which is critical in SLE pathogenesis. In this review, we discuss abnormal metabolic changes in glucose metabolism, tricarboxylic acid cycle, and lipid and amino acid metabolism as well as mitochondrial dysfunction in immune cells in SLE. We also review studies focused on the potential targets for key molecules of metabolic pathways in SLE, such as hypoxia-inducible factor-1α, mammalian target of rapamycin, and AMP-activated protein kinase. We highlight the therapeutic rationale for targeting these pathways in treating SLE and summarize their recent clinical applications in SLE.


Subject(s)
Lupus Erythematosus, Systemic , Humans , Inflammation , Signal Transduction
7.
Cell Mol Immunol ; 18(7): 1772-1782, 2021 07.
Article in English | MEDLINE | ID: mdl-32111985

ABSTRACT

Persistent hepatitis B virus (HBV) infection results in chronic liver diseases that may progress to chronic hepatitis, liver cirrhosis, and subsequent hepatocellular carcinoma. Previous studies demonstrated that adaptive immunity, in particular CD8 T cells, is critical in HBV elimination. Recent studies have revealed a distinct tissue-localized T cell lineage, tissue-resident memory (TRM) cells, that is crucial for protective immunity in peripheral tissues. In this study, we showed that treatment with an anti-asialo GM1 (ASGM1) antibody (Ab), which depletes NK cells, led to impairment of HBV clearance in a mouse animal model. Unexpectedly, the ability to clear HBV was not significantly impaired in NFIL3 KO mice, which are deficient in NK cells, implying that other non-NK ASGM1-positive immune cells mediate HBV clearance. We isolated intrahepatic ASGM1-positive cells from NFIL3 KO mice and analyzed the immune phenotype of these cells. Our results demonstrated a distinct population of CD44+ LFA-1hi CD8 T cells that were the major intrahepatic ASGM1-positive immune cells in NFIL3 KO mice. Importantly, transcriptome analysis revealed that these ASGM1-positive CD8 T cells had distinct gene profiles and shared a similar core gene signature with TRM cells. In addition to both transcriptional and phenotypic liver residency characteristics, ASGM1-positive CD8 T cells were able to home to and be retained in the liver after adoptive transfer. Taken together, our study results indicate that these ASGM1-positive liver-resident CD8 T cells are the major effector immune cells mediating anti-HBV immunity.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Animals , CD8-Positive T-Lymphocytes , G(M1) Ganglioside , Hepatitis B virus , Liver , Lymphocyte Function-Associated Antigen-1 , Mice , Mice, Inbred C57BL
8.
Rheumatology (Oxford) ; 59(11): 3340-3349, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32306043

ABSTRACT

OBJECTIVES: SLE is an autoimmune disease characterized by aberrant autoantibody production and immune dysfunctions. Whether the anti-CMV immunity is impaired in SLE patients is poorly understood. We investigated the specific anti-viral T-cell response in SLE patients with CMV infection and its possible impacts on clinical manifestations in lupus. METHODS: CD28 null T-cell percentages were measured by flow cytometry in 89 SLE patients and 58 healthy controls. A specific anti-CMV CD8 T-cell response was assessed ex vivo by the production of intracellular cytokines in response to CMV phosphoprotein 65 (pp65) by flow cytometry. Clinical manifestations and immune parameters were analysed in SLE patients according to their CMV serostatus. RESULTS: CD28 null T cells were significantly expanded in SLE patients. When the anti-CMV pp65 CD8 polyfunctional T cell response was analysed, as defined by production of at least three of four functional cytokines or effectors (intracellular IFN-γ, IL-2, TNF-α and surface CD107a), the results demonstrated that it was not impaired in SLE patients. In contrast, when comparing clinical manifestations, there were lower anti-ds-DNA levels and decreased SLEDAI in SLE patients with CMV infection. Furthermore, the expansion of CD4+CD28 null T cells was negatively associated with anti-ds-DNA levels and SLEDAI in these lupus patients. CONCLUSION: In SLE patients with CMV infection, the specific anti-CMV CD8 T-cell response is preserved but is associated with decreased disease activity and lower anti-DNA levels among these patients, suggesting CMV infection may mitigate lupus activity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Lupus Erythematosus, Systemic/immunology , Viral Matrix Proteins/immunology , Adult , Antibodies, Viral/blood , Antibody Specificity , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Case-Control Studies , Cytomegalovirus/immunology , Cytomegalovirus Infections/blood , DNA/immunology , Female , Flow Cytometry , Humans , Immunity, Cellular , Immunoglobulin G/blood , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/complications , Lymphocyte Activation , Lymphocytes, Null/immunology , Lysosomal-Associated Membrane Protein 1/biosynthesis , Male , Middle Aged , Tumor Necrosis Factor-alpha/biosynthesis
10.
Biochem Pharmacol ; 175: 113928, 2020 05.
Article in English | MEDLINE | ID: mdl-32217101

ABSTRACT

The cytokines interleukin-12 (IL-12) and IL-23 share a common IL-12/IL-23p40 subunit in structure and play a central role in T cell-mediated responses in inflammation. Over-activated IL-12 and IL-23 signaling drives aberrant T helper (Th) 1 and Th17 immune responses and contributes to immune-mediated diseases. Evidence from genome-wide association studies has shown that genetic alterations in the IL-12/IL-23 signaling pathways have significant links with chronic inflammation. In addition, accumulating evidence from animal models and clinical trials has provided insights into the effectiveness of blocking the IL-12/IL-23 pathways in immune regulation, broadening the clinical indications of IL-12/IL-23 pathway effectors in immune-mediated diseases. More recently, it has been addressed that the balance between IL and 12 and IL-23 is also critical in carcinogenesis. IL-12- and IL-23-driven T cell cytokines are especially important in controlling tumor initiation, growth, and metastasis, and thus, the IL-12/IL-23 pathway may be a promising target for immunotherapy. This review focuses on IL-12/IL-23 signal transduction and biological functionality in autoimmunity and oncoimmunology. We discuss the therapeutic rationale for targeting these cytokines to treat immune-mediated diseases and issues regarding their inadvertent consequences in the balance of host defense and tumor surveillance and summarize their recent clinical applications in immune-mediated diseases.


Subject(s)
Autoimmune Diseases/immunology , Interleukin-12/antagonists & inhibitors , Interleukin-12/immunology , Interleukin-23/antagonists & inhibitors , Interleukin-23/immunology , Neoplasms/immunology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/metabolism , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/immunology , Immunologic Factors/metabolism , Immunotherapy/methods , Immunotherapy/trends , Interleukin-12/metabolism , Interleukin-23/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
11.
J Cell Mol Med ; 24(2): 1822-1836, 2020 01.
Article in English | MEDLINE | ID: mdl-31821701

ABSTRACT

There is increasing evidence that statins, which are widely used in lowering serum cholesterol and the incidence of cardiovascular diseases, also exhibits anti-tumour properties. The underlying mechanisms by which statins-induced cancer cell death, however, remain incompletely understood. In this study, we explored the anti-tumour mechanisms of a lipophilic statin, lovastatin, in MCF-7 breast cancer cells. Lovastatin inhibited cell proliferation and induced cell apoptosis. Lovastatin caused p21 elevation while reduced cyclin D1 and survivin levels. Lovastatin also increased p53 phosphorylation, acetylation and its reporter activities. Results from chromatin immunoprecipitation analysis showed that p53 binding to the survivin promoter region was increased, while Sp1 binding to the region was decreased, in MCF-7 cells after lovastatin exposure. These actions were associated with liver kinase B1 (LKB1), AMP-activated protein kinase (AMPK) and p38 mitogen-activated protein kinase (p38MAPK) activation. Lovastatin's enhancing effects on p53 activation, p21 elevation and survivin reduction were significantly reduced in the presence of p38MAPK signalling inhibitor. Furthermore, LKB1-AMPK signalling blockade abrogated lovastatin-induced p38MAPK and p53 phosphorylation. Together these results suggest that lovastatin may activate LKB1-AMPK-p38MAPK-p53-survivin cascade to cause MCF-7 cell death. The present study establishes, at least in part, the signalling cascade by which lovastatin induces breast cancer cell death.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Lovastatin/pharmacology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Survivin/metabolism , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Apoptosis/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , MCF-7 Cells , Phosphorylation/drug effects , Signal Transduction/drug effects
12.
Cells ; 8(12)2019 12 13.
Article in English | MEDLINE | ID: mdl-31847240

ABSTRACT

Innate immune cells monitor invading pathogens and pose the first-line inflammatory response to coordinate with adaptive immunity for infection removal. Innate immunity also plays pivotal roles in injury-induced tissue remodeling and the maintenance of tissue homeostasis in physiological and pathological conditions. Lipid metabolites are emerging as the key players in the regulation of innate immune responses, and recent work has highlighted the importance of the lipid metabolite palmitate as an essential component in this regulation. Palmitate modulates innate immunity not only by regulating the activation of pattern recognition receptors in local innate immune cells, but also via coordinating immunological activity in inflammatory tissues. Moreover, protein palmitoylation controls various cellular physiological processes. Herein, we review the updated evidence that palmitate catabolism contributes to innate immune cell-mediated inflammatory processes that result in immunometabolic disorders.


Subject(s)
Fatty Acids/metabolism , Immunity, Innate/physiology , Inflammation/metabolism , Animals , Humans , Immunity, Innate/genetics , Palmitates/metabolism , Receptors, Pattern Recognition/metabolism
13.
Cells ; 8(9)2019 08 23.
Article in English | MEDLINE | ID: mdl-31450787

ABSTRACT

Type I and type III interferons (IFNs) share several properties in common, including the induction of signaling pathways, the activation of gene transcripts, and immune responses, against viral infection. Recent advances in the understanding of the molecular basis of innate and adaptive immunity have led to the re-examination of the role of these IFNs in autoimmune diseases. To date, a variety of IFN-regulated genes, termed IFN signature genes, have been identified. The expressions of these genes significantly increase in systemic lupus erythematosus (SLE), highlighting the role of type I and type III IFNs in the pathogenesis of SLE. In this review, we first discussed the signaling pathways and the immunoregulatory roles of type I and type III IFNs. Next, we discussed the roles of these IFNs in the pathogenesis of autoimmune diseases, including SLE. In SLE, IFN-stimulated genes induced by IFN signaling contribute to a positive feedback loop of autoimmunity, resulting in perpetual autoimmune inflammation. Based on this, we discussed the use of several specific IFN blocking strategies using anti-IFN-α antibodies, anti-IFN-α receptor antibodies, and IFN-α-kinoid or downstream small molecules, which intervene in Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathways, in clinical trials for SLE patients. Hopefully, the development of novel regimens targeting IFN signaling pathways will shed light on promising future therapeutic applications for SLE patients.


Subject(s)
Interferon Type I/metabolism , Interferons/metabolism , Lupus Erythematosus, Systemic/immunology , Adaptive Immunity/drug effects , Antibodies/pharmacology , Antibodies/therapeutic use , Gene Expression Regulation/drug effects , Humans , Immunity, Innate/drug effects , Lupus Erythematosus, Systemic/drug therapy , Molecular Targeted Therapy , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Interferon Lambda
14.
Cell Death Dis ; 10(2): 77, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30692521

ABSTRACT

Human osteoclast formation from mononuclear phagocyte precursors involves interactions between members of the tumor necrosis factor (TNF) ligand superfamily and their receptors. Recent evidence indicated that TNF-α-related apoptosis-inducing ligand (TRAIL) induces osteoclast differentiation via a TRAF6-dependent signaling pathway; but paradoxically, it inhibits RANK ligand (RANKL)-induced osteoclast differentiation. Although a number of signaling pathways were linked to the RANK and osteoclastogenesis, it is not known how TRAIL regulates RANK signaling. In this study, we demonstrate that TRAIL regulates RANK-induced osteoclastogenesis in terms of the assembly of lipid raft-associated signaling complexes. RANKL stimulation induced recruitment of TRAF6, c-Src, and DAP-12 into lipid rafts. However, the RANKL-induced assembly of lipid raft-associated signaling complexes and TRAF6 recruitment was abolished in the presence of TRAIL. TRAIL-induced dissociation of RANKL-induced lipid raft signaling complexes was reversed by treatment with TRAIL receptor (TRAIL-R) siRNA or an anti-TRAIL-R blocking antibody, indicating that TRAIL mediates suppression of RANKL-induced lipid raft signaling via interactions with TRAIL-R. Finally, we demonstrated that TRAIL suppressed inflammation-induced bone resorption and osteoclastogenesis in a collagen-induced arthritis (CIA) rat animal model. Our results provide a novel apoptosis-independent role of TRAIL in regulating RANK signaling and suppresses osteoclast activation via inhibiting lipid raft assembly and TRAF6 recruitment.


Subject(s)
Osteoclasts/metabolism , Peptide Fragments/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF Receptor-Associated Factor 6/genetics , Animals , Humans , Male , Rats , Rats, Sprague-Dawley , Signal Transduction , Transfection
15.
Mediators Inflamm ; 2018: 2403935, 2018.
Article in English | MEDLINE | ID: mdl-29670461

ABSTRACT

Spondyloarthropathy (SpA) is a unique type of joint inflammation characterized by coexisting erosive bone damage and pathological new bone formation. Previous genetic association studies have demonstrated that several cytokine pathways play a critical role in the pathogenesis of ankylosing spondylitis (AS), psoriatic arthritis (PsA), and other types of SpA. In addition to several well-known proinflammatory cytokines, recent studies suggest that IL-17 plays a pivotal role in the pathogenesis of SpA. Further evidence from human and animal studies have defined that IL-17 and IL-17-producing cells contribute to tissue inflammation, autoimmunity, and host defense, leading to the following pathologic events associated with SpA. Recently, several clinical trials targeting IL-17 pathways demonstrated the positive response of IL-17 blockade in treating AS, indicating a great potential of IL-17-targeting therapy in SpA. In this review article, we have discussed the contributing role of IL-17 and different IL-17-producing cells in the pathogenesis of SpA and provided an outline of therapeutic application of the IL-17 blockade in the treatment of SpA. Other targeted cytokines associated with IL-17 axis in SpA will also be included.


Subject(s)
Arthritis, Psoriatic/metabolism , Interleukin-17/metabolism , Spondylarthropathies/metabolism , Animals , Arthritis, Psoriatic/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-17/genetics , Spondylarthropathies/genetics , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/metabolism
16.
Front Immunol ; 9: 15, 2018.
Article in English | MEDLINE | ID: mdl-29403497

ABSTRACT

Objective: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces cell apoptosis by transducing apoptosis signals after interacting with its receptor (TRAIL-R). Although the actual biological role of TRAIL remains to be elucidated, recent accumulating evidence implies that TRAIL regulates immune responses and immune cell homeostasis via an apoptosis-independent pathway, suggesting a novel immune-regulatory role of TRAIL in autoimmune diseases. The purpose of this study is to address the immune-regulatory role and molecular mechanism of TRAIL in regulating T cell activation in autoimmune diseases. Design: TRAIL was administered to mice to induce experimental autoimmune encephalomyelitis (EAE), and to evaluate its impact on neuroinflammation and disease activity. The effects of TRAIL on neuroantigen [myelin oligodendrocyte glycoprotein (MOG)35-55]-activated T cell proliferation and cytokine production were investigated. TRAIL-treated MOG35-55-activated splenic Th17 cells were further adoptively transferred into Rag1 KO mice to induce passive EAE. Gene expression profiles of CD4+ T cells from EAE mice treated with TRAIL were analyzed by RNA sequencing and transcriptome analysis. Results: TRAIL suppressed autoimmune encephalomyelitis and inhibited T cell reactivity to neuro-antigen in murine EAE, and the effects were dependent on TRAIL-R signaling. Moreover, TRAIL directly inhibited activation of MOG35-55-activated CD4+ T cells, resulting in suppression of neuroinflammation and reduced disease activity in adoptive transfer-induced EAE. Furthermore, TRAIL-R signaling inhibited phosphorylation of proximal T cell receptor (TCR)-associated tyrosine kinases in activated CD4+ T cells. Importantly, TRAIL/TRAIL-R interaction downregulated TCR downstream signaling genes in RNA sequencing and transcriptome analysis. Conclusion: TRAIL/TRAIL-R interaction regulates CD4+ T cell activation in autoimmune inflammation and directly suppresses T cell activation via inhibiting TCR signaling, suggesting that TRAIL-R serves as a novel immune checkpoint in T cell responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Adoptive Transfer , Animals , Apoptosis/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/transplantation , Cell Proliferation , Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/metabolism , Peptide Fragments/metabolism , Signal Transduction/immunology
18.
J Formos Med Assoc ; 117(11): 1003-1010, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29174174

ABSTRACT

BACKGROUND/PURPOSE: Chronic periodontitis (CP) and rheumatoid arthritis (RA) are the most common chronic inflammatory diseases and their immunopathogenesis is similar. The aim of this study was to evaluate the effect of non-surgical periodontal treatment on the serum levels of RA-related inflammatory markers in patients with chronic periodontitis. METHODS: Thirty-one Taiwanese adults with CP were included. Demographics and periodontal parameters, including probing depth, clinical attachment level, and number of remaining teeth in the oral cavity, were recorded. All subjects received non-surgical periodontal treatment such as scaling and subgingival root planing. Serum samples were collected before and after the treatment. Serum levels of anti-citrullinated protein antibodies (ACPA), rheumatoid factor, tumor necrosis factor-α (TNF-α), C-reactive protein, interleukin-1ß (IL-1ß), and Interleukin-6 (IL-6) were measured using an enzyme-linked immunosorbent assay. RESULTS: Non-surgical periodontal treatment significantly reduced the serum ACPA (p = 0.015) and TNF-α levels (p = 0.026) in CP patients, particularly in patients with generalized CP. Furthermore, there was a significant and positive correlation between the number of extracted teeth and the reduction in the serum ACPA (p = 0.05) and IL-1ß levels (p = 0.029) after non-surgical periodontal treatment. CONCLUSION: Non-surgical periodontal therapy may aid in the control of RA-related inflammatory markers in patients with CP. A large-scale study with well-defined populations is needed to clarify the benefit of non-surgical periodontal therapy.


Subject(s)
Arthritis, Rheumatoid/blood , Biomarkers/blood , Chronic Periodontitis/blood , Chronic Periodontitis/therapy , Dental Scaling , Subgingival Curettage , Adult , Anti-Citrullinated Protein Antibodies/blood , Female , Humans , Interleukin-1beta/blood , Male , Middle Aged , Regression Analysis , Tumor Necrosis Factor-alpha/blood
19.
Cell Mol Immunol ; 15(9): 846-857, 2018 09.
Article in English | MEDLINE | ID: mdl-28392572

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has been implicated in the regulation of inflammation in rheumatoid arthritis (RA), primarily due to its ability to promote apoptosis in synoviocytes and infiltrating lymphocytes. The aim of this study was to investigate the immunomodulatory mechanism and role of TRAIL in inflammatory arthritis. We created an animal model of inflammatory arthritis and demonstrated that TRAIL significantly inhibited joint inflammation and reduced the severity of arthritis. The suppression of joint inflammation was not due to the TRAIL-mediated induction of apoptosis in T cells, macrophages or synovial fibroblasts. In contrast, TRAIL directly inhibited T-cell proliferation and suppressed the production of cytokines, which indicated that TRAIL exerted its anti-inflammatory effects by direct inhibition of T-cell activation. Moreover, TRAIL receptor (TRAIL-R)-knockout mice developed more severe disease, and the protective effects of TRAIL were abolished in the experimental arthritis model in TRAIL-R knockout mice. From these results, we conclude that TRAIL suppresses joint inflammation via an apoptosis-independent pathway and directly inhibits T-cell activation. Our results provide a novel apoptosis-independent, immune regulatory role for TRAIL in suppressing inflammatory arthritis and shed light on the development of effective new therapies for autoimmune inflammatory diseases.


Subject(s)
Arthritis, Experimental/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , TNF-Related Apoptosis-Inducing Ligand/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Cell Proliferation/genetics , Fibroblasts/immunology , Fibroblasts/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Synovial Membrane/immunology , Synovial Membrane/pathology , T-Lymphocytes/pathology , TNF-Related Apoptosis-Inducing Ligand/genetics
20.
J Mol Med (Berl) ; 94(5): 545-56, 2016 05.
Article in English | MEDLINE | ID: mdl-26631140

ABSTRACT

UNLABELLED: Galectin-3, a member of the ß-galactoside-binding lectin family, expresses in many different immune cells and modulates broad biological functions including cell adhesion, cell activation, cell growth, apoptosis, and inflammation. However, the role of galectin-3 in mucosal immunity or inflammatory bowel diseases is still not clear. We demonstrate here that galectin-3 knockout mice have more severe disease activity in the dextran sulfate sodium (DSS)-induced colitis model, indicating that galectin-3 may protect from inflammation in DSS-induced colitis. Furthermore, treating with galectin-3 reduced body weight loss, shortened colonic length, and ameliorated mucosal inflammation in mice having DSS-induced colitis. However, the protective effects of galectin-3 were eliminated by the administration of anti-CD25 mAb. In addition, primary T cells treated with galectin-3 ex vivo induced the expression of FOXP3, ICOS, and PD-1 with a Treg cell phenotype having a suppression function. Moreover, adoptive transfer of galectin-3-treated T cells reduced bowel inflammation and colitis in the T cell transfer colitis model. In conclusion, our results indicate that galectin-3 inhibited colonic mucosa inflammation and reduced disease severity by inducing regulatory T cells, suggesting that it is a potential therapeutic approach in inflammatory bowel disease. KEY MESSAGES: Galectin-3 offers protection from inflammation in experimental colitis. Galectin-3 knockout mice have more severe disease activity in DSS-induced colitis. Adoptive transfer of galectin-3-treated T cells reduced bowel inflammation. Galectin-3 inhibited colonic mucosa inflammation by inducing regulatory T cells. Galectin-3 is a potential therapeutic approach in inflammatory bowel disease.


Subject(s)
Colitis/etiology , Colitis/metabolism , Galectin 3/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Animals , Biomarkers , Colitis/drug therapy , Colitis/pathology , Dextran Sulfate/adverse effects , Disease Models, Animal , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Galectin 3/genetics , Galectin 3/pharmacology , Humans , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/metabolism , Intestinal Mucosa/drug effects , Lymphocyte Depletion , Mice , Mice, Knockout , Phenotype , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Severity of Illness Index , T-Lymphocyte Subsets , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...