Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 275
Filter
1.
Cytokine ; 176: 156540, 2024 04.
Article in English | MEDLINE | ID: mdl-38359559

ABSTRACT

Death Receptor 3 (DR3) is a cytokine receptor of the Tumor Necrosis Factor receptor superfamily that plays a multifaceted role in both innate and adaptive immunity. Based on the death domain motif in its cytosolic tail, DR3 had been proposed and functionally affirmed as a trigger of apoptosis. Further studies, however, also revealed roles of DR3 in other cellular pathways, including inflammation, survival, and proliferation. DR3 is expressed in various cell types, including T cells, B cells, innate lymphocytes, myeloid cells, fibroblasts, and even outside the immune system. Because DR3 is mainly expressed on T cells, DR3-mediated immune perturbations leading to autoimmunity and other diseases were mostly attributed to DR3 activation of T cells. However, which T cell subset and what T effector functions are controlled by DR3 to drive these processes remain incompletely understood. DR3 engagement was previously found to alter CD4 T helper subset differentiation, expand the Foxp3+ Treg cell pool, and maintain intraepithelial γδ T cells in the gut. Recent studies further unveiled a previously unacknowledged aspect of DR3 in regulating innate-like invariant NKT (iNKT) cell activation, expanding the scope of DR3-mediated immunity in T lineage cells. Importantly, in the context of iNKT cells, DR3 ligation exerted costimulatory effects in agonistic TCR signaling, unveiling a new regulatory framework in T cell activation and proliferation. The current review is aimed at summarizing such recent findings on the role of DR3 on conventional T cells and innate-like T cells and discussing them in the context of immunopathogenesis.


Subject(s)
Receptors, Cytokine , Receptors, Tumor Necrosis Factor, Member 25 , Humans , Tumor Necrosis Factor Ligand Superfamily Member 15 , Inflammation/metabolism , T-Lymphocyte Subsets/metabolism
2.
Proc Natl Acad Sci U S A ; 120(34): e2120771120, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37579137

ABSTRACT

The binding of tumor necrosis factor-like cytokine 1A (TL1A) to death receptor 3 (DR3) plays an important role in the interaction between dendritic cells (DCs) and T cells and contributes to intestinal inflammation development. However, the mechanism by which DCs expressing TL1A mediate helper T (Th) cell differentiation in the intestinal lamina propria (LP) during the pathogenesis of inflammatory bowel disease remains unclear. In this study, we found that TL1A/DR3 promoted Th1 and Th17 cell differentiation in T-T and DC-T cell interaction-dependent manners. TL1A-deficient CD4+ T cells failed to polarize into Th1/Th17 cells and did not cause colonic inflammation in a T cell transfer colitis model. Notably, TL1A was located in the cytoplasm and nuclei of DCs, positively regulated the DC-specific ICAM-grabbing nonintegrin/RAF1/nuclear factor κB signaling pathway, enhanced the antigen uptake ability of DCs, and promoted TLR4-mediated DC activation, inducing naive CD4+ T cell differentiation into Th1 and Th17 cells. Our work reveals that TL1A plays a regulatory role in inflammatory bowel disease pathogenesis.


Subject(s)
Inflammatory Bowel Diseases , Tumor Necrosis Factor Ligand Superfamily Member 15 , Humans , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Inflammatory Bowel Diseases/metabolism , Inflammation/metabolism , Tumor Necrosis Factor-alpha
3.
Respir Res ; 24(1): 182, 2023 Jul 11.
Article in English | MEDLINE | ID: mdl-37434162

ABSTRACT

Alveolar epithelial barrier is a potential therapeutic target for acute respiratory distress syndrome (ARDS). However, an effective intervention against alveolar epithelial barrier has not been developed. Here, based on single-cell RNA and mRNA sequencing results, death receptor 3 (DR3) and its only known ligand tumor necrosis factor ligand-associated molecule 1A (TL1A) were significantly reduced in epithelium from an ARDS mice and cell models. The apparent reduction in the TL1A/DR3 axis in lungs from septic-ARDS patients was correlated with the severity of the disease. The examination of knockout (KO) and alveolar epithelium conditional KO (CKO) mice showed that TL1A deficiency exacerbated alveolar inflammation and permeability in lipopolysaccharide (LPS)-induced ARDS. Mechanistically, TL1A deficiency decreased glycocalyx syndecan-1 and tight junction-associated zonula occludens 3 by increasing cathepsin E level for strengthening cell-to-cell permeability. Additionally, DR3 deletion aggravated barrier dysfunction and pulmonary edema in LPS-induced ARDS through the above mechanisms based on the analyses of DR3 CKO mice and DR3 overexpression cells. Therefore, the TL1A/DR3 axis has a potential value as a key therapeutic signaling for the protection of alveolar epithelial barrier.


Subject(s)
Receptors, Tumor Necrosis Factor, Member 25 , Respiratory Distress Syndrome , Tumor Necrosis Factor Ligand Superfamily Member 15 , Animals , Mice , Epithelium , Ligands , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/genetics , Tumor Necrosis Factor-alpha , Receptors, Tumor Necrosis Factor, Member 25/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics
4.
Cell Mol Life Sci ; 80(3): 76, 2023 Feb 27.
Article in English | MEDLINE | ID: mdl-36847849

ABSTRACT

Invariant natural killer T (iNKT) cells correspond to a population of thymus-generated T cells with innate-like characteristics and effector functions. Among the various iNKT subsets, NKT17 is the only subset that produces the proinflammatory cytokine IL-17. But, how NKT17 cells acquire this ability and what would selectively trigger their activation remain incompletely understood. Here, we identified the cytokine receptor DR3 being specifically expressed on thymic NKT17 cells and mostly absent on other thymic iNKT subsets. Moreover, DR3 ligation promoted the in vivo activation of thymic NKT17 cells and provided costimulatory effects upon agonistic α-GalCer stimulation. Thus, we identified a specific surface marker for thymic NKT17 cells that triggers their activation and augments their effector functions both in vivo and in vitro. These findings provide new insights for deciphering the role and function of murine NKT17 cells and for understanding the development and activation mechanisms of iNKT cells in general.


Subject(s)
Natural Killer T-Cells , Receptors, Tumor Necrosis Factor, Member 25 , Thymus Gland , Animals , Mice , Cytokines , Receptors, Cytokine , Receptors, Tumor Necrosis Factor, Member 25/metabolism
5.
Mol Neurobiol ; 60(1): 395-412, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36279100

ABSTRACT

Neuronal apoptosis is considered to be a critical cause of Alzheimer's disease (AD). Recently, meloxicam has shown neuroprotective effects; however, the inherent mechanisms are highly overlooked. Using APP/PS1 transgenic (Tg) mice as in vivo animal models, we found that meloxicam inhibits apoptosis in neurons by deactivating tumor necrosis factor receptor superfamily member 25 (TNFRSF25), leading to the suppression of the expression of fas-associated protein with death domain (FADD) and the cleavage of DNA fragmentation factor subunit α (DFFA) and cysteine aspartic acid protease-3 (caspase 3) via ß-amyloid protein (Aß)-depressing mechanisms. Moreover, the meloxicam treatment blocked the effects of ß-amyloid protein oligomers (Aßo) on stimulating the synthesis of tumor necrosis factor α (TNF-α) and TNF-like ligand 1A (TL1A) in neuroblastoma (N) 2a cells. TNF-α and TL1A induce apoptosis in neurons via TNFR- and TNFRSF25-dependent caspase 3-activating mechanisms, respectively. Knocking down the expression of TNFRSF25 blocked the effects of TL1A on inducing apoptosis in neurons by deactivating the signaling cascades of FADD, caspase 3, and DFFA. Consistently, TNFRSF25 shRNA blocked the effects of Aßo on inducing neuronal apoptosis, which was corroborated by the efficacy of meloxicam in inhibiting Aßo-induced neuronal apoptosis. By ameliorating neuronal apoptosis, meloxicam improved memory loss in APP/PS1 Tg mice.


Subject(s)
Alzheimer Disease , DNA Fragmentation , Meloxicam , Receptors, Tumor Necrosis Factor, Member 25 , Animals , Mice , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apoptosis , Caspase 3/metabolism , Meloxicam/pharmacology , Mice, Transgenic , Neurons/metabolism , Tumor Necrosis Factor-alpha/metabolism , Receptors, Tumor Necrosis Factor, Member 25/metabolism
6.
Front Immunol ; 13: 891328, 2022.
Article in English | MEDLINE | ID: mdl-35911746

ABSTRACT

TL1A, also called TNFSF15, is a member of tumor necrosis factor family. It is expressed in different immune cell, such as monocyte, macrophage, dendritic cell, T cell and non-immune cell, for example, synovial fibroblast, endothelial cell. TL1A competitively binds to death receptor 3 or decoy receptor 3, providing stimulatory signal for downstream signaling pathways, and then regulates proliferation, activation, apoptosis of and cytokine, chemokine production in effector cells. Recent findings showed that TL1A was abnormally expressed in autoimmune diseases, including rheumatoid arthritis, inflammatory bowel disease, psoriasis, primary biliary cirrhosis, systemic lupus erythematosus and ankylosing spondylitis. In vivo and in vitro studies further demonstrated that TL1A was involved in development and pathogenesis of these diseases. In this study, we comprehensively discussed the complex immunological function of TL1A and focused on recent findings of the pleiotropic activity conducted by TL1A in inflammatory autoimmune disease. Finish of the study will provide new ideas for developing therapeutic strategies for these diseases by targeting TL1A.


Subject(s)
Arthritis, Rheumatoid , Autoimmune Diseases , Inflammatory Bowel Diseases , Arthritis, Rheumatoid/complications , Autoimmune Diseases/complications , Humans , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism
7.
Front Immunol ; 13: 854995, 2022.
Article in English | MEDLINE | ID: mdl-35359966

ABSTRACT

Tumor necrosis factor (TNF)-like cytokine 1A (TL1A), a member of the TNF family, exists in the form of membrane-bound (mTL1A) and soluble protein (sTL1A). TL1A binding its only known functional receptor death domain receptor 3 (DR3) affects the transmission of various signals. This study first proposed that the TL1A/DR3 axis was significantly upregulated in patients and mice with both asthma and high TNF-a expression and in TNF-a-stimulated epithelial Beas-2B cells. Two independent approaches were used to demonstrate that the TL1A/DR3 axis of mice was strongly correlated with TNF-a in terms of exacerbating asthmatic epithelial-mesenchymal transformation (EMT). First, high expression levels of EMT proteins (e.g., collagen I, fibronectin, N-cadherin, and vimentin) and TL1A/DR3 axis were observed when mice airways were stimulated by recombinant mouse TNF-a protein. Moreover, EMT protein and TL1A/DR3 axis expression synchronously decreased after mice with OVA-induced asthma were treated with infliximab by neutralizing TNF-a activity. Furthermore, the OVA-induced EMT of asthmatic mice was remarkably improved upon the deletion of the TL1A/DR3 axis by knocking out the TL1A gene. TL1A siRNA remarkably intervened EMT formation induced by TNF-a in the Beas-2B cells. In addition, EMT was induced by the addition of high concentrations of recombinant human sTL1A with the cell medium. The TL1A overexpression via pc-mTL1A in vitro remarkably increased the EMT formation induced by TNF-a. Overall, these findings indicate that the TL1A/DR3 axis may have a therapeutic role for asthmatic with high TNF-a level.


Subject(s)
Asthma , Receptors, Tumor Necrosis Factor, Member 25 , Animals , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Humans , Mice , Ovalbumin , Receptors, Tumor Necrosis Factor, Member 25/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism
8.
Am J Pathol ; 192(4): 722-736, 2022 04.
Article in English | MEDLINE | ID: mdl-35063404

ABSTRACT

Similar to the behavior of inflamed tubular epithelial cells, clear cell renal cell carcinoma (ccRCC) cells express death receptor 3 (DR3 or TNFSFR25) in situ, and expression increases with tumor grade. Surprisingly, E-selectin, which can be induced in endothelial cells by DR3 signaling, is also expressed by ccRCC cells and increases with tumor grade. In ccRCC organ cultures, addition of tumor necrosis factor-like 1A (TL1A or TNFSF15), the ligand for DR3, activates NF-κB and mitogen-activated protein kinases, induces both DR3 and E-selectin expression in an NF-κB-dependent manner, and promotes cell cycle entry. DR3 immunoprecipitated from ccRCC tissue contains sialyl Lewis X moieties (the ligand recognized by E-selectin), proximity ligation assays reveal DR3, and E-selectin interacts on ccRCC cells. Similar to that with the addition of TL1A, the addition of soluble E-selectin to ccRCC organ cultures activates NF-κB and mitogen-activated protein kinases in ccRCC cells and increases both DR3 and E-selectin expression and cell-cycle entry. In contrast, normal renal tubular epithelium, which poorly expresses DR3, is minimally responsive to either of these ligands. These data suggest a functional role for autocrine/paracrine DR3/E-selectin interactions in ccRCC and its progression, revealing a potential new target for therapeutic intervention.


Subject(s)
Carcinoma, Renal Cell , E-Selectin , Kidney Neoplasms , Receptors, Tumor Necrosis Factor, Member 25 , Antigens, CD , Carcinoma, Renal Cell/metabolism , E-Selectin/genetics , E-Selectin/metabolism , Endothelial Cells/metabolism , Female , Humans , Kidney Neoplasms/metabolism , Ligands , Male , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor, Member 25/genetics , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism
9.
Prostate ; 82(1): 154-166, 2022 01.
Article in English | MEDLINE | ID: mdl-34662447

ABSTRACT

BACKGROUND: Metabolic reprograming is now a recognized hallmark of cancer. The prostate-specific phosphatase and tensin homolog deleted on chromosome 10 (Pten) gene-conditional knockout (KO) mouse carcinogenesis model is highly desirable for studying prostate cancer biology and prevention due to its close resemblance of primary molecular defects and histopathological features of human prostate cancer. We have recently published macromolecular profiling of this model by proteomics and transcriptomics, denoting a preeminence of inflammation and myeloid suppressive immune cell features. Here, we performed metabolomic analyses of Pten-KO prostate versus wild type (WT) counterpart for discernable changes in the aqueous metabolites and contrasted to those in the TRAMP neuroendocrine carcinoma (NECa). METHODS: Three matched pairs of tissue-specific conditional Pten-KO mouse prostate and WT prostate of litter/cage-mates at 20-22 weeks of age and three pairs of TRAMP NECa versus WT (28-31 weeks) were profiled for their global aqueous metabolite changes, using hydrophilic interaction liquid chromatography-tandem mass spectrometry. RESULTS: The Pten-KO prostate increased purine nucleotide pools, cystathionine, and both reduced and oxidized glutathione (GSH, GSSG), and gluconate/glucuronate species in addition to cholesteryl sulfate and polyamine precursor ornithine. On the contrary, Pten-KO prostate contained diminished pools of glycolytic intermediates and phosphorylcholine derivatives, select amino acids, and their metabolites. Bioinformatic integration revealed a significant shunting of glucose away from glycolysis-citrate cycle and glycerol-lipid genesis to pentose phosphate cycle for NADPH/GSH/GSSG redox and pentose moieties for purine and pyrimidine nucleotides, and glycosylation/glucuronidation. Implicit arginine catabolism to ornithine was consistent with immunosuppression in Pten-KO model. While also increased in cystathionine-GSH/GSSG, purine, and pyrimidine nucleotide pools and glucuronidation at the expense of glycolysis-citrate cycle, the TRAMP NECa increased abundance of many amino acids, methyl donor S-adenosyl-methionine, and intermediates for phospholipids without increasing cholesteryl sulfate or ornithine. CONCLUSIONS: The aqueous metabolomic patterns in Pten-KO prostate and TRAMP NECa shared similarities in the greater pools of cystathionine, GSH/GSSG redox pair, and nucleotides and shunting away from glycolysis-citrate cycle in both models. Remarkable metabolic distinctions between them included metabolisms of many amino acids (protein synthesis; arginine-ornithine/immune suppression) and cholesteryl sulfate and methylation donor for epigenetic regulations.


Subject(s)
Carcinoma, Neuroendocrine , PTEN Phosphohydrolase/metabolism , Prostate , Prostatic Neoplasms , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Animals , Biomarkers/analysis , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Chromatography, Liquid/methods , Disease Models, Animal , Male , Metabolomics/methods , Mice , Mice, Knockout , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tandem Mass Spectrometry/methods
10.
Cells ; 10(11)2021 11 10.
Article in English | MEDLINE | ID: mdl-34831323

ABSTRACT

Signaling via death receptor family members such as TNF-R1 mediates pleiotropic biological outcomes ranging from inflammation and proliferation to cell death. Pro-survival signaling is mediated via TNF-R1 complex I at the cellular plasma membrane. Cell death induction requires complex IIa/b or necrosome formation, which occurs in the cytoplasm. In many cell types, full apoptotic or necroptotic cell death induction requires the internalization of TNF-R1 and receptosome formation to properly relay the signal inside the cell. We interrogated the role of the enzyme A disintegrin and metalloprotease 17 (ADAM17)/TACE (TNF-α converting enzyme) in death receptor signaling in human hematopoietic cells, using pharmacological inhibition and genetic ablation. We show that in U937 and Jurkat cells the absence of ADAM17 does not abrogate, but rather increases TNF mediated cell death. Likewise, cell death triggered via DR3 is enhanced in U937 cells lacking ADAM17. We identified ADAM17 as the key molecule that fine-tunes death receptor signaling. A better understanding of cell fate decisions made via the receptors of the TNF-R1 superfamily may enable us, in the future, to more efficiently treat infectious and inflammatory diseases or cancer.


Subject(s)
ADAM17 Protein/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , ADAM17 Protein/antagonists & inhibitors , ADAM17 Protein/deficiency , Amyloid Precursor Protein Secretases/metabolism , Cell Death , Cell Survival , Endocytosis , Humans , Jurkat Cells , MCF-7 Cells , Models, Biological , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
11.
Front Immunol ; 12: 692127, 2021.
Article in English | MEDLINE | ID: mdl-34305924

ABSTRACT

Mucus secretion is an important feature of asthma that highly correlates with morbidity. Current therapies, including administration of mucolytics and anti-inflammatory drugs, show limited effectiveness and durability, underscoring the need for novel effective and longer lasting therapeutic approaches. Here we show that mucus production in the lungs is regulated by the TNF superfamily member 15 (TL1A) acting through the mucus-inducing cytokine IL-13. TL1A induces IL13 expression by innate lymphoid cells leading to mucus production, in addition to promoting airway inflammation and fibrosis. Reciprocally, neutralization of IL13 signaling through its receptor (IL4Rα), completely reverses TL1A-induced mucus secretion, while maintaining airway inflammation and fibrosis. Importance of TL1A is further demonstrated using a preclinical asthma model induced by chronic house dust mite exposure where TL1A neutralization by genetic deletion or antagonistic blockade of its receptor DR3 protected against mucus production and fibrosis. Thus, TL1A presents a promising therapeutic target that out benefits IL13 in reversing mucus production, airway inflammation and fibrosis, cardinal features of severe asthma in humans.


Subject(s)
Asthma/immunology , Interleukin-13/immunology , Interleukin-4 Receptor alpha Subunit/immunology , Lung/immunology , Mucus/immunology , Receptors, Tumor Necrosis Factor, Member 25/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Animals , Asthma/pathology , DNA-Binding Proteins/genetics , Female , Fibrosis , Lung/pathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Member 25/genetics , Signal Transduction
12.
Carcinogenesis ; 42(5): 772-783, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33710266

ABSTRACT

Prostate cancer (PCa) constitutes a serious health challenge and remains one of the main causes of cancer-related death among men. The more aggressive form of the disease has been attributed to androgen independence, resulting in a lack of response to androgen deprivation therapy and sustained activation of other growth pathways. The scaffold proteins ß-arrestin 1 and 2 (ßarr1 and ßarr2), which are known to mediate G protein-coupled receptor desensitization and internalization, were also shown to modulate prostate tumorigenesis. ßarr1 is significantly overexpressed (>4-fold) in PCa cells relative to ßarr2. In this study, we investigated the effect of ßarr1 overexpression in PCa development and progression using the mouse and human PCa cell xenografts, and autochthonous transgenic adenocarcinoma of the mouse prostate (TRAMP) models deficient in ß-arrestin depletion of ßarr1 in TRAMP mice (TRAMP/ßarr1-/-) increased PCa growth and decreased overall survival relative to control TRAMP or TRAMP/ßarr2-/- animals. Prostate tissues from TRAMP/ßarr1-/- tumors displayed an increase in androgen receptor (AR) expression, whereas overexpression of ßarr1 in TRAMP-C1 (TRAMP-C1-ßarr1-GFP) which derived from TRAMP decreased AR expression, cell proliferation and tumor growth in nude mice xenografts, relative to control TRAMP-C1-GFP. Knockdown of ßarr1 expression in human MDA PCa 2b cells (MDA PCa 2b-ßarr1-/-) also decreased AR expression cell proliferation and tumor growth relative to control (MDA PCa 2b-Sham) cells. Interestingly, both TRAMP-C1-ßarr1-GFP and MDA PCa 2b-ßarr1-/- xenografts showed a decrease in AKT phosphorylation but an increase in MAPK activation. Altogether, the data indicate that the effect of ßarr1 in modulating AR signaling to regulate PCa aggressiveness is cell and host autonomous.


Subject(s)
Carcinogenesis/genetics , Prostatic Neoplasms/genetics , Receptors, Tumor Necrosis Factor, Member 25/genetics , beta-Arrestin 1/genetics , beta-Arrestin 2/genetics , Animals , Animals, Genetically Modified , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Male , Mice , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Signal Transduction
13.
Front Immunol ; 12: 636789, 2021.
Article in English | MEDLINE | ID: mdl-33737937

ABSTRACT

Corneal transplantation (CT) is the most frequent type of solid organ transplant (SOT) performed worldwide. Unfortunately, immunological rejection is the primary cause of graft failure for CT and therefore advances in immune regulation to induce tolerance remains an unmet medical need. Recently, our work and others in pre-clinical studies found that cyclophosphamide (Cy) administered after ("post-transplant," PTCy) hematopoietic stem cell transplantation (HSCT), i.e., liquid transplants is effective for graft vs. host disease prophylaxis and enhances overall survival. Importantly, within the past 10 years, PTCy has been widely adopted for clinical HSCT and the results at many centers have been extremely encouraging. The present studies found that Cy can be effectively employed to prolong the survival of SOT, specifically mouse corneal allografts. The results demonstrated that the timing of PTCy administration is critical for these CT and distinct from the kinetics employed following allogeneic HSCT. PTCy was observed to interfere with neovascularization, a process critically associated with immune rejection of corneal tissue that ensues following the loss of ocular "immune privilege." PTCy has the potential to delete or directly suppress allo-reactive T cells and treatment here was shown to diminish T cell rejection responses. These PTCy doses were observed to spare significant levels of CD4+ FoxP3+ (Tregs) which were found to be functional and could readily receive stimulating signals leading to their in vivo expansion via TNFRSF25 and CD25 agonists. In total, we posit future studies can take advantage of Cy based platforms to generate combinatorial strategies for long-term tolerance induction.


Subject(s)
Corneal Transplantation , Cyclophosphamide/therapeutic use , Graft Rejection/prevention & control , Postoperative Complications/prevention & control , Allografts/immunology , Animals , Cells, Cultured , Forkhead Transcription Factors/genetics , Graft Rejection/etiology , Humans , Immune Tolerance , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Signal Transduction
14.
Dis Markers ; 2021: 8858905, 2021.
Article in English | MEDLINE | ID: mdl-33628338

ABSTRACT

Epigenetic gene silencing by aberrant DNA methylation leads to loss of key cellular pathways in tumorigenesis. In order to analyze the effects of DNA methylation on prostate cancer, we established LNCaP-derived human prostate cancer cells that can pharmacologically induce global reactivation of hypermethylated genes by the methyl-CpG targeted transcriptional activation (MeTA) method. The MeTA suppressed the growth of LNCaP-derived cells and induced apoptosis. Microarray analysis indicated that PYCARD (PYD and CARD domain containing) encoding an apoptosis-inducing factor was upregulated by 65-fold or more after treatment with MeTA. We analyzed DNA methylation statuses using 50 microdissected primary prostate cancer tissues and found an extremely high frequency of tumor-specific promoter hypermethylation of PYCARD (90%, 45/50). Moreover, DNA methylation status was significantly associated with Gleason score (P = 0.0063); the frequency of tumor-specific hypermethylation was 96% (44/46) in tumors with Gleason score ≥ 7, whereas that in tumors with Gleason score 6 was 25% (1/4). Immunohistochemical analyses using these 50 cases indicated that only 8% (4/50) of cancerous tissues expressed PYCARD, whereas 80% (40/50) of corresponding normal prostate epithelial and/or basal cells expressed PYCARD. In addition, there was no relationship between PYCARD immunostaining and the Gleason score in cancerous tissue and surrounding normal tissue. Inducible expression of PYCARD inhibited cell proliferation by induction of apoptosis. These results suggest that aberrant methylation of PYCARD is a distinctive feature of prostate cancers with Gleason score ≥ 7 and may play an important role in escaping from apoptosis in prostatic tumorigenesis.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , Carcinogenesis/genetics , DNA Methylation , Epigenesis, Genetic , Prostatic Neoplasms/genetics , Aged , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line , Cell Line, Tumor , CpG Islands , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neoplasm Grading , Promoter Regions, Genetic , Prostate/metabolism , Prostate/pathology , Prostatectomy/methods , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Receptors, Tumor Necrosis Factor, Member 25/genetics , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Signal Transduction
15.
Cell Mol Gastroenterol Hepatol ; 11(1): 249-272, 2021.
Article in English | MEDLINE | ID: mdl-32827707

ABSTRACT

BACKGROUND & AIMS: TNFSF15 genetic variants leading to increased TNF superfamily member 15 (TNFSF15) expression confer risk for inflammatory bowel disease (IBD), and TNFSF15 is being explored as a therapeutic target in IBD patients. Although the focus for TNFSF15-mediated inflammatory outcomes has been predominantly on its action on T cells, TNFSF15 also promotes inflammatory outcomes in human macrophages. Given the critical role for macrophages in bacterial clearance, we hypothesized that TNFSF15 promotes antimicrobial pathways in human macrophages and that macrophages from TNFSF15 IBD risk carriers with higher TNFSF15 expression have an advantage in these antimicrobial outcomes. METHODS: We analyzed protein expression, signaling, bacterial uptake, and intracellular bacterial clearance in human monocyte-derived macrophages through flow cytometry, enzyme-linked immunosorbent assay, and gentamicin protection. RESULTS: Autocrine/paracrine TNFSF15 interactions with death receptor 3 (DR3) were required for optimal levels of pattern-recognition-receptor (PRR)-induced bacterial clearance in human macrophages. TNFSF15 induced pyruvate dehydrogenase kinase 1-dependent bacterial uptake and promoted intracellular bacterial clearance through reactive oxygen species, nitric oxide synthase 2, and autophagy up-regulation. The TNFSF15-initiated TNF receptor-associated factor 2/receptor-interacting protein kinase 1/RIP3 pathway was required for mitogen-activated protein kinase and nuclear factor-κB activation, and, in turn, induction of each of the antimicrobial pathways; the TNFSF15-initiated Fas-associated protein with death domain/mucosa-associated lymphoid tissue lymphoma translocation protein 1/caspase-8 pathway played a less prominent role in antimicrobial functions, despite its key role in TNFSF15-induced cytokine secretion. Complementation of signaling pathways or antimicrobial pathways restored bacterial uptake and clearance in PRR-stimulated macrophages where TNFSF15:DR3 interactions were inhibited. Monocyte-derived macrophages from high TNFSF15-expressing rs6478108 TT IBD risk carriers in the TNFSF15 region showed increased levels of the identified antimicrobial pathways. CONCLUSIONS: We identify that autocrine/paracrine TNFSF15 is required for optimal PRR-enhanced antimicrobial pathways in macrophages, define mechanisms regulating TNFSF15-dependent bacterial clearance, and determine how the TNFSF15 IBD risk genotype modulates these outcomes.


Subject(s)
Inflammatory Bowel Diseases/immunology , Macrophages/immunology , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Animals , Autocrine Communication/immunology , Cells, Cultured , Enterococcus faecalis/immunology , Enterococcus faecalis/isolation & purification , Escherichia coli/immunology , Escherichia coli/isolation & purification , Genetic Predisposition to Disease , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Macrophages/metabolism , Mice , Paracrine Communication/immunology , Primary Cell Culture , Receptors, Pattern Recognition/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics
16.
J Matern Fetal Neonatal Med ; 34(18): 2965-2970, 2021 Sep.
Article in English | MEDLINE | ID: mdl-31608721

ABSTRACT

INTRODUCTION: Preeclampsia has the highest rate of obstetric morbidity and mortality. METHODS: We recruited 21 women with preeclampsia and 27 women with uncomplicated pregnancies. We used a quantitative protein macroarray that allowed for analysis of 40 proteins. RESULTS: We found a statistically significant increase in the concentration of DR3, LIF and a significant decrease of VEGF, PlGF, syndecan-4 and galectin-2, in the plasma of women with preeclampsia. CONCLUSIONS: There are no previous studies assessing syndecan 4, galectin 2, and DR3 concentrations in women with preeclampsia; Our results indicate these proteins are new factors that play important roles in the immunological pathomechanism of preeclampsia.


Subject(s)
Galectin 2/genetics , Pre-Eclampsia , Receptors, Tumor Necrosis Factor, Member 25/genetics , Syndecan-4/genetics , Biomarkers , Female , Humans , Placenta Growth Factor , Pregnancy
17.
Int J Radiat Oncol Biol Phys ; 109(5): 1547-1558, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33188861

ABSTRACT

PURPOSE: To investigate the temporal and spatial infiltration of TRAMP-C1 tumors by myeloid-derived suppressor cells (MDSCs) after high-dose radiation therapy (RT), and to explore their effect on tumor growth. METHODS AND MATERIALS: TRAMP-C1 intramuscularly tumors were irradiated with a single dose of 8 Gy or 25 Gy. The dynamics of infiltrated MDSCs and their intratumoral spatial distribution were assessed by immunohistochemistry and flow cytometry. Cytokine levels in the blood and tumor were analyzed by multiplex immunoassay. Mice were injected with anti-Gr-1 antibody to determine whether MDSCs affect tumor growth after RT. RESULTS: CD11b+Gr-1+ MDSCs infiltrated TRAMP-C1 tumors irradiated with 25 Gy, but not 8 Gy, within 4 hours and recruitment persisted for at least 2 weeks. Both CD11b+Ly6G+Ly6C+ polymorphonuclear-MDSCs (PMN-MDSCs) and CD11b+Ly6G-Ly6Chi monocytic-MDSCs (M-MDSCs) were involved. Tumor RT also increased the representation of both MDSC subpopulations in the spleen and peripheral blood. Levels of multiple cytokines were increased in the tumors at 2 weeks, including GM-CSF, G-CSF, CCL-3, CCL-5, CXCL-5, IL-6, IL-17α, and VEGF-a; while G-CSF, IL-6, and TNF-α levels increased in the blood. PMN-MDSCs aggregated in the central necrotic region of the irradiated tumors over time, where they were associated with avascular hypoxia (CD31-PIMO+). MDSCs expressed the proangiogenic factor, matrix metalloproteinase-9, and, within the necrotic area, high levels of arginase-1 and indoleamine 2,3-dioxygenase. Depletion of PMN-MDSCs by Gr-1 antibody increased the efficacy of high-dose RT. CONCLUSIONS: PMN-MDSCs infiltrate TRAMP-C1 tumors after high-dose RT. Their spatial distribution suggests they are involved in the evolution of an intratumoral state of necrosis associated with avascular hypoxia, and their phenotype is consistent with them being immunosuppressive. They appear to promote tumor growth after RT, making them a prime therapeutic target for therapeutic intervention. Assessment of MDSCs and cytokine levels in blood could be an index of the need for such an intervention.


Subject(s)
Myeloid-Derived Suppressor Cells/physiology , Prostatic Neoplasms/radiotherapy , Receptors, Tumor Necrosis Factor, Member 25 , Animals , CD11b Antigen , Cell Movement , Chemokines/analysis , Cytokines/analysis , Disease Models, Animal , Flow Cytometry , Immunoassay/methods , Male , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/radiation effects , Prostatic Neoplasms/blood , Prostatic Neoplasms/immunology , Radiotherapy Dosage , Receptors, Chemokine/immunology , Tumor Microenvironment
18.
J Immunol ; 205(12): 3348-3357, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33177161

ABSTRACT

Relative control of HIV-1 infection has been linked to genetic and immune host factors. In this study, we analyzed 96 plasma proteome arrays from chronic untreated HIV-1-infected individuals using the classificatory random forest approach to discriminate between uncontrolled disease (plasma viral load [pVL] >50,000 RNA copies/ml; CD4 counts 283 cells/mm3, n = 47) and relatively controlled disease (pVL <10,000 RNA copies/ml; CD4 counts 657 cells/mm3, n = 49). Our analysis highlighted the TNF molecule's relevance, in particular, TL1A (TNFSF15) and its cognate DR3 (TNFSRF25), both of which increased in the relative virus control phenotype. DR3 levels (in plasma and PBMCs) were validated in unrelated cohorts (including long-term nonprogressors), thus confirming their independence from CD4 counts and pVL. Further analysis in combined antiretroviral treatment (cART)-treated individuals with a wide range of CD4 counts (137-1835 cells/mm3) indicated that neither TL1A nor DR3 levels reflected recovery of CD4 counts with cART. Interestingly, in cART-treated individuals, plasma TL1A levels correlated with regulatory T cell frequencies, whereas soluble DR3 was strongly associated with the abundance of effector HLA-DR+CD8+ T cells. A positive correlation was also observed between plasma DR3 levels and the HIV-1-specific T cell responses. In vitro, costimulation of PBMC with DR3-specific mAb increased the magnitude of HIV-1-specific responses. Finally, in splenocytes of DNA.HTI-vaccinated mice, costimulation of HTI peptides and a DR3 agonist (4C12) intensified the magnitude of T cell responses by 27%. These data describe the role of the TL1A-DR3 axis in the natural control of HIV-1 infection and point to the use of DR3 agonists in HIV-1 vaccine regimens.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Receptors, Tumor Necrosis Factor, Member 25/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/metabolism , Female , HIV Infections/blood , HIV-1/metabolism , Humans , Male , Mice , Receptors, Tumor Necrosis Factor, Member 25/blood , Tumor Necrosis Factor Ligand Superfamily Member 15/blood
19.
Sci Rep ; 10(1): 18189, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097818

ABSTRACT

Tumor necrosis factor-like cytokine 1A (TL1A, TNFSF15) is implicated in inflammatory bowel disease, modulating the location and severity of inflammation and fibrosis. TL1A expression is increased in inflamed mucosa and associated with fibrostenosing Crohn's disease. Tl1a-overexpression in mice causes spontaneous ileitis, and exacerbates induced proximal colitis and fibrosis. Intestinal fibroblasts express Death-receptor 3 (DR3; the only know receptor for TL1A) and stimulation with TL1A induces activation in vitro. However, the contribution of direct TL1A-DR3 activation on fibroblasts to fibrosis in vivo remains unknown. TL1A overexpressing naïve T cells were transferred into Rag-/- , Rag-/- mice lacking DR3 in all cell types (Rag-/-Dr3-/-), or Rag-/- mice lacking DR3 only on fibroblasts (Rag-/-Dr3∆Col1a2) to induce colitis and fibrosis, assessed by clinical disease activity index, intestinal inflammation, and collagen deposition. Rag-/- mice developed overt colitis with intestinal fibrostenosis. In contrast, Rag-/-Dr3-/- demonstrated decreased inflammation and fibrosis. Despite similar clinical disease and inflammation as Rag-/-, Rag-/-Dr3∆Col1a2 exhibited reduced intestinal fibrosis and attenuated fibroblast activation and migration. RNA-Sequencing of TL1A-stimulated fibroblasts identified Rho signal transduction as a major pathway activated by TL1A and inhibition of this pathway modulated TL1A-mediated fibroblast functions. Thus, direct TL1A signaling on fibroblasts promotes intestinal fibrosis in vivo. These results provide novel insight into profibrotic pathways mediated by TL1A paralleling its pro-inflammatory effects.


Subject(s)
Intestinal Diseases/metabolism , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Animals , Fibroblasts/metabolism , Fibrosis/metabolism , Mice , Mice, Transgenic , Receptors, Tumor Necrosis Factor, Member 25/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics
20.
Nat Commun ; 11(1): 4718, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32948777

ABSTRACT

Disturbances in glucose homeostasis and low-grade chronic inflammation culminate into metabolic syndrome that increase the risk for the development of type 2 diabetes mellitus (T2DM). The recently discovered group 2 innate lymphoid cells (ILC2s) are capable of secreting copious amounts of type 2 cytokines to modulate metabolic homeostasis in adipose tissue. In this study, we have established that expression of Death Receptor 3 (DR3), a member of the TNF superfamily, on visceral adipose tissue (VAT)-derived murine and peripheral blood human ILC2s is inducible by IL-33. We demonstrate that DR3 engages the canonical and/or non-canonical NF-κB pathways, and thus stimulates naïve and co-stimulates IL-33-activated ILC2s. Importantly, DR3 engagement on ILC2s significantly ameliorates glucose tolerance, protects against insulin-resistance onset and remarkably reverses already established insulin-resistance. Taken together, these results convey the potent role of DR3 as an ILC2 regulator and introduce DR3 agonistic treatment as a novel therapeutic avenue for treating T2DM.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/metabolism , Lymphocytes/metabolism , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Adipocytes/metabolism , Adolescent , Adult , Aged , Animals , Cytokines/metabolism , DNA-Binding Proteins/genetics , Female , Glucose/metabolism , Homeostasis , Humans , Immunity, Innate , Insulin Resistance , Interleukin-33/metabolism , Intra-Abdominal Fat/metabolism , Male , Metabolic Syndrome/complications , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Receptors, Tumor Necrosis Factor, Member 25/therapeutic use , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...