Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nature ; 588(7836): 151-156, 2020 12.
Article in English | MEDLINE | ID: mdl-33149305

ABSTRACT

Lymphotoxin ß-receptor (LTßR) signalling promotes lymphoid neogenesis and the development of tertiary lymphoid structures1,2, which are associated with severe chronic inflammatory diseases that span several organ systems3-6. How LTßR signalling drives chronic tissue damage particularly in the lung, the mechanism(s) that regulate this process, and whether LTßR blockade might be of therapeutic value have remained unclear. Here we demonstrate increased expression of LTßR ligands in adaptive and innate immune cells, enhanced non-canonical NF-κB signalling, and enriched LTßR target gene expression in lung epithelial cells from patients with smoking-associated chronic obstructive pulmonary disease (COPD) and from mice chronically exposed to cigarette smoke. Therapeutic inhibition of LTßR signalling in young and aged mice disrupted smoking-related inducible bronchus-associated lymphoid tissue, induced regeneration of lung tissue, and reverted airway fibrosis and systemic muscle wasting. Mechanistically, blockade of LTßR signalling dampened epithelial non-canonical activation of NF-κB, reduced TGFß signalling in airways, and induced regeneration by preventing epithelial cell death and activating WNT/ß-catenin signalling in alveolar epithelial progenitor cells. These findings suggest that inhibition of LTßR signalling represents a viable therapeutic option that combines prevention of tertiary lymphoid structures1 and inhibition of apoptosis with tissue-regenerative strategies.


Subject(s)
Lung/drug effects , Lung/physiology , Lymphotoxin beta Receptor/antagonists & inhibitors , Regeneration/drug effects , Signal Transduction/drug effects , Wnt Proteins/agonists , Adaptive Immunity , Aging/metabolism , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Apoptosis/drug effects , Emphysema/metabolism , Female , Humans , Immunity, Innate , Lung/metabolism , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Smoke/adverse effects , Stem Cells/drug effects , Stem Cells/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
2.
Cell Commun Signal ; 17(1): 171, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31878945

ABSTRACT

BACKGROUND: Lymphotoxin ß receptor (LTßR) plays important roles in the development of the immune system and immune response. At the cellular level, ligand-bound LTßR activates the pro-inflammatory NF-κB pathway but the detailed mechanisms regulating its signaling remain unknown. Understanding them is of high importance since LTßR and its ligands are promising therapeutic targets. Here, we studied the consequences of perturbed cellular cholesterol content on LTßR-induced NF-κB signaling. METHODS: To modulate cholesterol availability and/or level in lung carcinoma A549 and H2228, and endothelial HUVEC cells different treatment regimens with filipin, methyl-ß-cyclodextrin and simvastatin were applied. LTßR localization was studied by confocal microscopy. The activity of LTßR-induced NF-κB pathway was assessed by measuring the levels of NF-κB pathway inhibitor IκBα and phosphorylation of RelA transcription factor by Western blotting. The NF-κB transcriptional response, production of chemokines and adhesion molecules were examined by qRT-PCR, ELISA, and Western blotting, respectively. Adherence of different types of primary immune cells to epithelial A549 cells and endothelial HUVECs was measured fluorometrically. Interactions of LTßR with its protein partners were investigated by immunoprecipitation. RESULTS: We showed that filipin-mediated sequestration of cholesterol or its depletion from the plasma membrane with methyl-ß-cyclodextrin impaired LTßR internalization and potentiated LTßR-dependent activation of the canonical branch of the NF-κB pathway. The latter was manifested by enhanced degradation of IκBα inhibitor, elevated RelA phosphorylation, substantial increase in the expression of NF-κB target genes encoding, among others, cytokines and adhesion molecules known to play important roles in immune response. It was followed by robust secretion of CXCL8 and upregulation of ICAM1, that favored the adhesion of immune cells (NK and T cells, neutrophils) to A549 cells and HUVECs. Mechanistically, we showed that cholesterol depletion stabilized interactions of ligand-stimulated LTßR with modified forms of TRAF2 and NEMO proteins. CONCLUSIONS: Our results showed that the reduction of the plasma membrane content of cholesterol or its sequestration strongly potentiated signaling outcome initiated by LTßR. Thus, drugs modulating cholesterol levels could potentially improve efficacy of LTßR-based therapies. Video abstract.


Subject(s)
Cholesterol/pharmacology , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , A549 Cells , Cells, Cultured , Humans , Jurkat Cells , Neutrophils/drug effects , Neutrophils/metabolism
3.
Sci Signal ; 11(541)2018 07 31.
Article in English | MEDLINE | ID: mdl-30065029

ABSTRACT

Members of the family of nuclear factor κB (NF-κB) transcription factors are critical for multiple cellular processes, including regulating innate and adaptive immune responses, cell proliferation, and cell survival. Canonical NF-κB complexes are retained in the cytoplasm by the inhibitory protein IκBα, whereas noncanonical NF-κB complexes are retained by p100. Although activation of canonical NF-κB signaling through the IκBα kinase complex is well studied, few regulators of the NF-κB-inducing kinase (NIK)-dependent processing of noncanonical p100 to p52 and the subsequent nuclear translocation of p52 have been identified. We discovered a role for cyclin-dependent kinase 12 (CDK12) in transcriptionally regulating the noncanonical NF-κB pathway. High-content phenotypic screening identified the compound 919278 as a specific inhibitor of the lymphotoxin ß receptor (LTßR), and tumor necrosis factor (TNF) receptor superfamily member 12A (FN14)-dependent nuclear translocation of p52, but not of the TNF-α receptor-mediated nuclear translocation of p65. Chemoproteomics identified CDK12 as the target of 919278. CDK12 inhibition by 919278, the CDK inhibitor THZ1, or siRNA-mediated knockdown resulted in similar global transcriptional changes and prevented the LTßR- and FN14-dependent expression of MAP3K14 (which encodes NIK) as well as NIK accumulation by reducing phosphorylation of the carboxyl-terminal domain of RNA polymerase II. By coupling a phenotypic screen with chemoproteomics, we identified a pathway for the activation of the noncanonical NF-κB pathway that could serve as a therapeutic target in autoimmunity and cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic , NF-kappa B/metabolism , Osteosarcoma/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Cyclins/metabolism , Gene Expression Profiling , High-Throughput Screening Assays , Humans , Indoles/pharmacology , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Propionates/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteome , Signal Transduction , TWEAK Receptor/antagonists & inhibitors , TWEAK Receptor/genetics , TWEAK Receptor/metabolism , Tumor Cells, Cultured , NF-kappaB-Inducing Kinase
4.
Gut ; 66(8): 1369-1381, 2017 08.
Article in English | MEDLINE | ID: mdl-27196595

ABSTRACT

OBJECTIVE: Lymphotoxin ß receptor (LTßR) signalling has been implicated in inflammation-associated tumour development in different tissues. We have analysed the role of LTßR and alternative NF-κB signalling in Helicobacter pylori-mediated gastric inflammation and pathology. DESIGN: We analysed several ligands and receptors of the alternative NF-κB pathway, RelB, p52 nuclear translocation and target genes in tissue samples of H. pylori-infected patients with different degrees of gastritis or early gastric tumours by in situ hybridisation, immunohistochemistry, Western blot and real-time PCR analyses. Molecular mechanisms involved in LTßR activation by H. pylori were assessed in vitro using human gastric cancer cell lines and distinct H. pylori isolates. The effects of blocking or agonistically activating LTßR on gastric pathology during challenge with a human pathogenic H. pylori strain were studied in a mouse model. RESULTS: Among the tested candidates, LT was significantly increased and activated alternative NF-κB signalling was observed in the gastric mucosa of H. pylori-infected patients. H. pyloriinduced LTßR-ligand expression in a type IV secretion system-dependent but CagA-independent manner, resulting in activation of the alternative NF-κB pathway, which was further enhanced by blocking canonical NF-κB during infection. Blocking LTßR signalling in vivo suppressed H. pylori-driven gastritis, whereas LTßR activation in gastric epithelial cells of infected mice induced a broadened pro-inflammatory chemokine milieu, resulting in exacerbated pathology. CONCLUSIONS: LTßR-triggered activation of alternative NF-κB signalling in gastric epithelial cells executes H. pylori-induced chronic gastritis, representing a novel target to restrict gastric inflammation and pathology elicited by H. pylori, while exclusively targeting canonical NF-κB may aggravate pathology by enhancing the alternative pathway.


Subject(s)
Chemokines/metabolism , Gastritis/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori , Lymphotoxin beta Receptor/metabolism , NF-kappa B/metabolism , Type IV Secretion Systems/metabolism , Animals , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Line, Tumor , Chemokine CCL2/metabolism , Chemokine CCL20/metabolism , Chemokine CXCL10/metabolism , Epithelial Cells/metabolism , Female , Gastric Mucosa/metabolism , Gastritis/microbiology , Helicobacter Infections/complications , Humans , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/genetics , Mice , Mice, Inbred C57BL , RNA, Messenger , Signal Transduction , Transcription Factor RelB/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Tumor Necrosis Factor-alpha/pharmacology
5.
Kidney Int ; 89(1): 113-26, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26398497

ABSTRACT

Accumulation of inflammatory cells in different renal compartments is a hallmark of progressive kidney diseases including glomerulonephritis (GN). Lymphotoxin ß receptor (LTßR) signaling is crucial for the formation of lymphoid tissue, and inhibition of LTßR signaling has ameliorated several non-renal inflammatory models. Therefore, we tested whether LTßR signaling could also have a role in renal injury. Renal biopsies from patients with GN were found to express both LTα and LTß ligands, as well as LTßR. The LTßR protein and mRNA were localized to tubular epithelial cells, parietal epithelial cells, crescents, and cells of the glomerular tuft, whereas LTß was found on lymphocytes and tubular epithelial cells. Human tubular epithelial cells, mesangial cells, and mouse parietal epithelial cells expressed both LTα and LTß mRNA upon stimulation with TNF in vitro. Several chemokine mRNAs and proteins were expressed in response to LTßR signaling. Importantly, in a murine lupus model, LTßR blockade improved renal function without the reduction of serum autoantibody titers or glomerular immune complex deposition. Thus, a preclinical mouse model and human studies strongly suggest that LTßR signaling is involved in renal injury and may be a suitable therapeutic target in renal diseases.


Subject(s)
Glomerulonephritis, IGA/metabolism , Lupus Nephritis/metabolism , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/metabolism , RNA, Messenger/analysis , Signal Transduction , Adult , Animals , Cell Line , Chemokines/genetics , Chemokines/metabolism , Disease Models, Animal , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Female , Glomerulonephritis, IGA/genetics , Humans , Immunoglobulins/pharmacology , Kidney Glomerulus/chemistry , Kidney Glomerulus/pathology , Kidney Tubules/chemistry , Kidney Tubules/metabolism , Kidney Tubules/pathology , Ligands , Lupus Nephritis/genetics , Lymphocytes/chemistry , Lymphotoxin beta Receptor/analysis , Lymphotoxin beta Receptor/genetics , Lymphotoxin-alpha/analysis , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Lymphotoxin-beta/analysis , Lymphotoxin-beta/genetics , Lymphotoxin-beta/metabolism , Male , Mesangial Cells/metabolism , Mice , Middle Aged , Signal Transduction/drug effects , Transcriptome
6.
Cytokine Growth Factor Rev ; 25(2): 139-45, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24698108

ABSTRACT

The Lymphotoxin (LT) pathway is best known for its role in orchestrating the development and homeostasis of lymph nodes and Peyer's patches through the regulation of homeostatic chemokines. More recently an appreciation of the LTßR pathway in the production of Type I interferons (IFN-I) during homeostasis and infection has emerged. LTßR signaling is essential in differentiating stromal cells and macrophages in lymphoid organs to rapidly produce IFN-I in response to virus infections independently of the conventional TLR signaling systems. In addition, LTßR signaling is required to produce homeostatic levels of IFN-I from dendritic cells in order to effectively cross-prime a CD8+ T cell response to protein antigen. Importantly, pharmacological inhibition of LTßR signaling in mice has a profound positive impact on a number of autoimmune disease models, although it remains unclear if this efficacy is linked to IFN-I production during chronic inflammation. In this review, we will provide a brief overview of how the "Lymphotoxin Network" is linked to the IFN-I response and its impact on the immune system.


Subject(s)
Autoimmune Diseases/drug therapy , Interferon Type I/immunology , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/immunology , Lymphotoxin-alpha/immunology , Adaptive Immunity , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Inflammation/immunology , Interferon Type I/biosynthesis , Macrophages/immunology , Mice , Signal Transduction/immunology
7.
EMBO Rep ; 15(5): 540-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24668263

ABSTRACT

A receptor-ligand interaction can evoke a broad range of biological activities in different cell types depending on receptor identity and cell type-specific post-receptor signaling intermediates. Here, we show that the TNF family member LIGHT, known to act as a death-triggering factor in motoneurons through LT-ßR, can also promote axon outgrowth and branching in motoneurons through the same receptor. LIGHT-induced axonal elongation and branching require ERK and caspase-9 pathways. This distinct response involves a compartment-specific activation of LIGHT signals, with somatic activation-inducing death, while axonal stimulation promotes axon elongation and branching in motoneurons. Following peripheral nerve damage, LIGHT increases at the lesion site through expression by invading B lymphocytes, and genetic deletion of Light significantly delays functional recovery. We propose that a central and peripheral activation of the LIGHT pathway elicits different functional responses in motoneurons.


Subject(s)
Axons/physiology , Motor Neurons/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Animals , B-Lymphocytes/immunology , Butadienes/pharmacology , Caspase 9/metabolism , Caspase Inhibitors/pharmacology , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Knockout , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Tumor Necrosis Factor Ligand Superfamily Member 14/biosynthesis
8.
Science ; 343(6176): 1221-8, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24557838

ABSTRACT

Current antiviral agents can control but not eliminate hepatitis B virus (HBV), because HBV establishes a stable nuclear covalently closed circular DNA (cccDNA). Interferon-α treatment can clear HBV but is limited by systemic side effects. We describe how interferon-α can induce specific degradation of the nuclear viral DNA without hepatotoxicity and propose lymphotoxin-ß receptor activation as a therapeutic alternative. Interferon-α and lymphotoxin-ß receptor activation up-regulated APOBEC3A and APOBEC3B cytidine deaminases, respectively, in HBV-infected cells, primary hepatocytes, and human liver needle biopsies. HBV core protein mediated the interaction with nuclear cccDNA, resulting in cytidine deamination, apurinic/apyrimidinic site formation, and finally cccDNA degradation that prevented HBV reactivation. Genomic DNA was not affected. Thus, inducing nuclear deaminases-for example, by lymphotoxin-ß receptor activation-allows the development of new therapeutics that, in combination with existing antivirals, may cure hepatitis B.


Subject(s)
Antiviral Agents/pharmacology , DNA, Circular/metabolism , DNA, Viral/metabolism , Hepatitis B virus/drug effects , Hepatitis B/drug therapy , Hepatocytes/drug effects , Interferon-alpha/pharmacology , Lymphotoxin beta Receptor/agonists , Animals , Antibodies, Monoclonal , Antiviral Agents/therapeutic use , Cell Line , Cell Nucleus/virology , Cytidine/metabolism , Cytidine Deaminase/biosynthesis , Hepatitis B virus/metabolism , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Interferon-alpha/therapeutic use , Liver/drug effects , Liver/metabolism , Liver/virology , Lymphotoxin beta Receptor/antagonists & inhibitors , Mice, SCID , Minor Histocompatibility Antigens , Proteins , Up-Regulation
9.
Am J Transplant ; 13(3): 541-51, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23356438

ABSTRACT

The exchange of information during interactions of T cells with dendritic cells, B cells or other T cells regulates the course of T, B and DC-cell activation and their differentiation into effector cells. The tumor necrosis factor superfamily member LIGHT (homologous to lymphotoxin, exhibits inducible expression and competes with HSV glycoprotein D for binding to herpesvirus entry mediator, a receptor expressed on T lymphocytes) is transiently expressed upon T cell activation and modulates CD8 T cell-mediated alloreactive responses upon herpes virus entry mediator (HVEM) and lymphotoxin ß receptor (LTßR) engagement. LIGHT-deficient mice, or WT mice treated with LIGHT-targeting decoy receptors HVEM-Ig, LTßR-Ig or sDcR3-Ig, exhibit prolonged graft survival compared to untreated controls, suggesting that LIGHT modulates the course and severity of graft rejection. Therefore, targeting the interaction of LIGHT with HVEM and/or LTßR using recombinant soluble decoy receptors or monoclonal antibodies represent an innovative therapeutic strategy for the prevention and treatment of allograft rejection and for the promotion of donor-specific tolerance.


Subject(s)
Antibodies, Monoclonal/pharmacology , Graft Rejection/immunology , Graft vs Host Disease/immunology , Lymphotoxin beta Receptor/antagonists & inhibitors , Organ Transplantation , Tumor Necrosis Factor Ligand Superfamily Member 14/antagonists & inhibitors , Animals , Humans , Lymphotoxin beta Receptor/immunology , Lymphotoxin beta Receptor/metabolism , Mice , Protein Binding , Transplantation, Homologous , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology
10.
Mol Immunol ; 54(1): 40-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23220069

ABSTRACT

Our earlier studies indicated that activation of the lymphotoxin beta receptor (LTßR) by T cell derived LTα(1)ß(2) regulates inflammatory cytokine expression. While characterizing the cellular and molecular mechanisms responsible for the down regulation of the inflammatory reaction after LTßR stimulation we were able to identify the specific induction of TRIM30α expression as a result of LTßR signalling in mouse macrophages. Furthermore, we could demonstrate that LTßR activation in these cells results in the down regulation of pro-inflammatory cytokine (e.g. TNF and IL-6) and mediator expression upon TLR4 and TLR9 re-stimulation, demonstrating that LTßR activation on mouse macrophages dampens pro-inflammatory cytokine and mediator expression. Thus, LTßR signalling renders macrophages hypo-responsive to subsequent stimulation with TLR ligands. The observation of an LTßR-mediated TLR-tolerance in the human monocyte cell line THP-1 suggests that similar signalling mechanisms seem to exist in human cells. Signalling pathway analysis clearly demonstrated that LTßR-induced TRIM30α expression is mediated by an IκBα-dependent signalling pathway. Furthermore, the LTßR-induced TRIM30α expression seems to be TRAF3 dependent. Our data suggest that LTßR activation on mouse macrophages is involved in the control of pro-inflammatory cytokine and mediator expression by activation of a signalling pathway that controls exacerbating inflammatory cytokine production.


Subject(s)
Cytokines/genetics , Intracellular Signaling Peptides and Proteins/physiology , Lymphotoxin beta Receptor/physiology , TNF Receptor-Associated Factor 3/physiology , Animals , Antibodies, Monoclonal/pharmacology , Cells, Cultured , Cytokines/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lymphotoxin beta Receptor/agonists , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/physiology , Mice , Signal Transduction/genetics , Signal Transduction/immunology , Signal Transduction/physiology , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism
11.
Am J Transplant ; 12(9): 2322-34, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22594431

ABSTRACT

The lymphotoxin system (LT) regulates interactions between lymphocytes and stromal cells to maintain lymphoid microenvironmental homeostasis. Soluble LT beta-receptor-Ig (LTßRIg) blocks lymphocyte LTα1ß2-stromal cell LTßR signaling. In a murine cardiac allograft model, LTbRIg treatment reversed the tolerance induced by anti-CD40L antibody leading to graft inflammation and fibrosis. LTßRIg treatment decreased PD-L1 expression by blood endothelial cells, and decreased VCAM-1 while increasing CXCL1, CXCL2, CXCL12, CCL5, CCL21 and IL-6 expression in fibroblastic reticular cells. In secondary lymphoid organs these effects caused T- and B cell zone disruption, loss of CD35(+) follicular dendritic cells and abnormal recruitment of CD11b(+) Ly6G(+) neutrophils. These disruptions correlated with increased numbers of CD8(+) T cells and CD11b(+) Ly6G(+) neutrophils, and decreased numbers of CD4(+) T cells and Foxp3(+) regulatory T cells in the grafts. Depleting neutrophils or blocking neutrophil-attracting chemokines restored normal histology in lymph node, spleen and grafts. Taken together, LTßRIg treatment altered stromal subset, particularly fibroblastic reticular cell, production of cytokines and chemokines, resulting in changes in neutrophil recruitment in spleen, lymph node and grafts, and inflammation and fibrosis associated with decreased Foxp3(+) regulatory T cells and increased CD8(+) T cell infiltration of grafts.


Subject(s)
Fibrosis/etiology , Heart Transplantation , Inflammation/etiology , Lymphotoxin beta Receptor/antagonists & inhibitors , Animals , Chemokines/metabolism , Immunoglobulins/immunology , Immunohistochemistry , Lymphotoxin beta Receptor/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/immunology
12.
Arthritis Res Ther ; 13(6): R182, 2011.
Article in English | MEDLINE | ID: mdl-22044682

ABSTRACT

INTRODUCTION: In Sjögren's syndrome, keratoconjunctivitis sicca (dry eye) is associated with infiltration of lacrimal glands by leukocytes and consequent losses of tear-fluid production and the integrity of the ocular surface. We investigated the effect of blockade of the lymphotoxin-beta receptor (LTBR) pathway on lacrimal-gland pathology in the NOD mouse model of Sjögren's syndrome. METHODS: Male NOD mice were treated for up to ten weeks with an antagonist, LTBR-Ig, or control mouse antibody MOPC-21. Extra-orbital lacrimal glands were analyzed by immunohistochemistry for high endothelial venules (HEV), by Affymetrix gene-array analysis and real-time PCR for differential gene expression, and by ELISA for CXCL13 protein. Leukocytes from lacrimal glands were analyzed by flow-cytometry. Tear-fluid secretion-rates were measured and the integrity of the ocular surface was scored using slit-lamp microscopy and fluorescein isothiocyanate (FITC) staining. The chemokine CXCL13 was measured by ELISA in sera from Sjögren's syndrome patients (n = 27) and healthy controls (n = 30). Statistical analysis was by the two-tailed, unpaired T-test, or the Mann-Whitney-test for ocular integrity scores. RESULTS: LTBR blockade for eight weeks reduced B-cell accumulation (approximately 5-fold), eliminated HEV in lacrimal glands, and reduced the entry rate of lymphocytes into lacrimal glands. Affymetrix-chip analysis revealed numerous changes in mRNA expression due to LTBR blockade, including reduction of homeostatic chemokine expression. The reduction of CXCL13, CCL21, CCL19 mRNA and the HEV-associated gene GLYCAM-1 was confirmed by PCR analysis. CXCL13 protein increased with disease progression in lacrimal-gland homogenates, but after LTBR blockade for 8 weeks, CXCL13 was reduced approximately 6-fold to 8.4 pg/mg (+/- 2.7) from 51 pg/mg (+/-5.3) in lacrimal glands of 16 week old control mice. Mice given LTBR blockade exhibited an approximately two-fold greater tear-fluid secretion than control mice (P = 0.001), and had a significantly improved ocular surface integrity score (P = 0.005). The mean CXCL13 concentration in sera from Sjögren's patients (n = 27) was 170 pg/ml, compared to 92.0 pg/ml for sera from (n = 30) healthy controls (P = 0.01). CONCLUSIONS: Blockade of LTBR pathways may have therapeutic potential for treatment of Sjögren's syndrome.


Subject(s)
Chemokine CXCL13/metabolism , Cornea/metabolism , Lacrimal Apparatus/metabolism , Lymphotoxin beta Receptor/metabolism , Sjogren's Syndrome/metabolism , Adult , Aged , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Chemokine CXCL13/genetics , Cornea/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Female , Gene Expression/drug effects , Gene Expression Profiling , Humans , Immunohistochemistry , Keratoconjunctivitis Sicca/drug therapy , Keratoconjunctivitis Sicca/genetics , Keratoconjunctivitis Sicca/metabolism , Lacrimal Apparatus/drug effects , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/immunology , Male , Mice , Mice, Inbred NOD , Microscopy, Fluorescence , Middle Aged , Mucins/genetics , Mucins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sjogren's Syndrome/drug therapy , Sjogren's Syndrome/genetics , Tears/metabolism , Venules/metabolism , Venules/physiology
13.
Biochem Biophys Res Commun ; 404(4): 1060-4, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21195057

ABSTRACT

Adiponectin exerts anti-diabetic and anti-atherogenesis properties through its 2 receptors (AdipoR1 and AdipoR2). However, the signaling pathways responsible for the anti-inflammatory effects of adiponectin are largely unknown. In this study, we identified the lymphotoxin (LT)-ß receptor (LTBR) as an interacting partner of human AdipoR1 by using a yeast two-hybrid screening. The interaction between LTBR and AdipoR1 was confirmed by co-immunoprecipitation and co-localization analysis. Furthermore, adiponectin incubation inhibited lymphotoxin-induced NF-κB activation and the expression of adhesion molecules in human umbilical vein endothelial cells. These results indicated that AdipoR1 interacted with LTBR and mediated the inhibition of LTBR-activated NF-κB pathway.


Subject(s)
Adiponectin/metabolism , Endothelium, Vascular/metabolism , Lymphotoxin beta Receptor/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Receptors, Adiponectin/metabolism , Animals , COS Cells , Chlorocebus aethiops , Humans , Immunoprecipitation , Lymphotoxin alpha1, beta2 Heterotrimer/metabolism , Lymphotoxin beta Receptor/metabolism , NF-kappa B/metabolism , Two-Hybrid System Techniques , Umbilical Veins/cytology
14.
Arthritis Res Ther ; 11(1): R24, 2009.
Article in English | MEDLINE | ID: mdl-19222863

ABSTRACT

INTRODUCTION: The lymphotoxin-beta receptor (LTbetaR) pathway is important in the development and maintenance of lymphoid structures. Blocking this pathway has proven beneficial in murine models of autoimmune diseases such as diabetes and rheumatoid arthritis. The aim of this study was to determine the effects of LTbetaR pathway blockade on Sjögren syndrome (SS)-like salivary gland disease in non-obese diabetic (NOD) mice. METHODS: The course of SS-like disease was followed in NOD mice that were given lymphotoxin-beta receptor-immunoglobulin fusion protein (LTbetaR-Ig) starting at 9 weeks of age. Treatment was given as a single weekly dose for 3, 7, or 10 weeks. Age-matched NOD mice treated with mouse monoclonal IgG1, or not treated at all, were used as controls. The severity of inflammation, cellular composition, and lymphoid neogenesis in the submandibular glands were determined by immunohistochemistry. Mandibular lymph nodes were also studied. Saliva flow rates were measured, and saliva was analyzed by a multiplex cytokine assay. The salivary glands were analyzed for CXCL13, CCL19, and CCL21 gene expression by quantitative polymerase chain reaction. RESULTS: Treatment with LTbetaR-Ig prevented the increase in size and number of focal infiltrates normally observed in this SS-like disease. Compared with the controls, the submandibular glands of LTbetaR-Ig-treated mice had fewer and smaller T- and B-cell zones and fewer high endothelial venules per given salivary gland area. Follicular dendritic cell networks were lost in LTbetaR-Ig-treated mice. CCL19 expression was also dramatically inhibited in the salivary gland infiltrates. Draining lymph nodes showed more gradual changes after LTbetaR-Ig treatment. Saliva flow was partially restored in mice treated with 10 LTbetaR-Ig weekly injections, and the saliva cytokine profile of these mice resembled that of mice in the pre-disease state. CONCLUSIONS: Our findings show that blocking the LTbetaR pathway results in ablation of the lymphoid organization in the NOD salivary glands and thus an improvement in salivary gland function.


Subject(s)
Lymphotoxin beta Receptor/immunology , Salivary Glands/immunology , Salivary Glands/pathology , Signal Transduction/immunology , Sjogren's Syndrome/immunology , Sjogren's Syndrome/pathology , Animals , Chemokine CCL19/biosynthesis , Chemokine CCL21/biosynthesis , Chemokine CXCL13/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Immunohistochemistry , Lymphotoxin beta Receptor/antagonists & inhibitors , Mice , Mice, Inbred NOD , Reverse Transcriptase Polymerase Chain Reaction
15.
J Exp Med ; 206(1): 233-48, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19139167

ABSTRACT

Atherosclerosis involves a macrophage-rich inflammation in the aortic intima. It is increasingly recognized that this intimal inflammation is paralleled over time by a distinct inflammatory reaction in adjacent adventitia. Though cross talk between the coordinated inflammatory foci in the intima and the adventitia seems implicit, the mechanism(s) underlying their communication is unclear. Here, using detailed imaging analysis, microarray analyses, laser-capture microdissection, adoptive lymphocyte transfers, and functional blocking studies, we undertook to identify this mechanism. We show that in aged apoE(-/-) mice, medial smooth muscle cells (SMCs) beneath intimal plaques in abdominal aortae become activated through lymphotoxin beta receptor (LTbetaR) to express the lymphorganogenic chemokines CXCL13 and CCL21. These signals in turn trigger the development of elaborate bona fide adventitial aortic tertiary lymphoid organs (ATLOs) containing functional conduit meshworks, germinal centers within B cell follicles, clusters of plasma cells, high endothelial venules (HEVs) in T cell areas, and a high proportion of T regulatory cells. Treatment of apoE(-/-) mice with LTbetaR-Ig to interrupt LTbetaR signaling in SMCs strongly reduced HEV abundance, CXCL13, and CCL21 expression, and disrupted the structure and maintenance of ATLOs. Thus, the LTbetaR pathway has a major role in shaping the immunological characteristics and overall integrity of the arterial wall.


Subject(s)
Aorta, Abdominal/growth & development , Apolipoproteins E/genetics , Connective Tissue/growth & development , Lymphotoxin beta Receptor/physiology , Signal Transduction/physiology , Aging , Animals , Aorta, Abdominal/metabolism , Atherosclerosis/genetics , Biological Transport , Cells, Cultured , Chemokine CCL21/genetics , Chemokine CCL21/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Cluster Analysis , Connective Tissue/metabolism , Gene Expression Profiling , In Situ Hybridization , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/growth & development , Lymphoid Tissue/metabolism , Lymphotoxin beta Receptor/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Organogenesis , Reverse Transcriptase Polymerase Chain Reaction , Tunica Intima/growth & development , Tunica Intima/metabolism , Tunica Media/growth & development , Tunica Media/metabolism
16.
Mol Immunol ; 45(1): 34-41, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17590442

ABSTRACT

The lymphotoxin beta receptor (LTbetaR) signalling pathway is involved in the development of secondary lymphoid organs and the maintenance of organized lymphoid tissues. Additionally, previous studies clearly demonstrated the involvement of the LTbetaR interaction with its ligands in promoting intestinal inflammation. In order to dissect the role of LTbetaR activation in the mouse model of acute DSS-induced colitis we treated mice with a functional inhibitor of LTbetaR activation (LTbetaR:Ig) and compared it to disease in LTbetaR-deficient and LTalphabeta-deficient mice. All these modes of LTbetaR signalling ablation resulted in significant aggravation of the disease and in release of inflammatory cytokines such as TNF, IL-6, and IFNgamma. Finally, using mice with conditionally ablated expression of membrane bound LTbeta on T or B cells, respectively, distinct and opposite contributions of surface LTbeta expressed on T or B cells was found. Thus, activation of LTbetaR by LTalphabeta mainly expressed on T lymphocytes is crucial for the down regulation of the inflammatory response in this experimental model.


Subject(s)
B-Lymphocytes/metabolism , Colitis/metabolism , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin-beta/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Acute Disease , Animals , B-Lymphocytes/drug effects , Colitis/chemically induced , Colitis/enzymology , Colitis/pathology , Disease Models, Animal , Female , Immunoglobulins/pharmacology , Inflammation/metabolism , Ligands , Lymphotoxin beta Receptor/deficiency , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Inbred C57BL , Peroxidase/metabolism , Signal Transduction/drug effects , T-Lymphocytes/drug effects , Weight Loss/drug effects
17.
J Immunol ; 179(10): 6901-9, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17982081

ABSTRACT

Although studies indicate LIGHT (lymphotoxin (LT)-like, exhibits inducible expression and competes with HSV glycoprotein D for herpes virus entry mediator (HVEM), a receptor expressed by T lymphocytes) enhances inflammation and T cell-mediated immunity, the mechanisms involved in this process remain obscure. In this study, we assessed the role of LIGHT in IL-12 production and development of CD4(+) Th cells type one (Th1) in vivo. Bone marrow-derived dendritic cells from LIGHT(-/-) mice were severely impaired in IL-12p40 production following IFN-gamma and LPS stimulation in vitro. Furthermore, blockade of LIGHT in vitro and in vivo with HVEM-Ig and LT beta receptor (LTbetaR)-Ig leads to impaired IL-12 production and defective polyclonal and Ag-specific IFN-gamma production in vivo. In an infection model, injection of HVEM-Ig or LTbetaR-Ig into the usually resistant C57BL/6 mice results in defective IL-12 and IFN-gamma production and severe susceptibility to Leishmania major that was reversed by rIL-12 treatment. This striking susceptibility to L. major in mice injected with HVEM-Ig or LTbetaR-Ig was also reproduced in LIGHT(-/-) --> RAG1(-/-) chimeric mice. In contrast, L. major-infected LTbeta(-/-) mice do not develop acute disease, suggesting that the effect of LTbetaR-Ig is not due to blockade of membrane LT (LTalpha1beta2) signaling. Collectively, our data show that LIGHT plays a critical role for optimal IL-12 production by DC and the development of IFN-gamma-producing CD4(+) Th1 cells and its blockade results in severe susceptibility to Leishmania major.


Subject(s)
Immunity, Innate , Interleukin-12 Subunit p40/immunology , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Receptors, Tumor Necrosis Factor, Member 14/immunology , Th1 Cells/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Animals , Dendritic Cells/immunology , Disease Models, Animal , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Immunity, Cellular/drug effects , Immunity, Cellular/genetics , Immunity, Innate/drug effects , Immunity, Innate/genetics , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Leishmaniasis, Cutaneous/genetics , Lipopolysaccharides/pharmacology , Lymphotoxin alpha1, beta2 Heterotrimer , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/immunology , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor, Member 14/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 14/genetics , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
18.
JPEN J Parenter Enteral Nutr ; 31(5): 358-64; discussion 364-5, 2007.
Article in English | MEDLINE | ID: mdl-17712143

ABSTRACT

BACKGROUND: Mucosal addressin cellular adhesion molecule-1 (MAdCAM-1) directs lymphocyte migration into gut-associated lymphoid tissue (GALT) through Peyer's patches (PPs). Parenteral nutrition (PN) impairs mucosal immunity by reducing PPs MAdCAM-1 expression, T and B cells in GALT, and intestinal and respiratory immunoglobulin (Ig) A levels. We previously showed that PN reduces lymphotoxin beta receptor blockade (LTbetaR) in PPs and intestine, and that stimulation with LTbetaR agonist antibodies reverses these defects. To confirm that LTbetaR regulates transcription of MAdCAM-1 message and more fully understand the effects of LTbetaR on MAdCAM-1 function within the mucosal immune system, we studied the effect of LTbetaR blockade with a chimeric LTbetaR Ig-fusion protein on MAdCAM-1 mRNA levels, PP lymphocyte mass and IgA levels in the intestinal and respiratory tracts. METHODS: Mice were cannulated and killed 3 days after receiving chow + control Ig, chow + LTbetaR-Ig fusion protein (100 microg IV), or PN + control Ig. The PPs of half of the animals were processed for lymphocyte count, and the other half were processed for complementary DNA and subsequent polymerase chain reaction (PCR). mRNA levels of MAdCAM-1 were determined by real-time PCR; intestinal and respiratory IgA levels were measured by ELISA. RESULTS: PN significantly reduced PP lymphocyte mass, MAdCAM-1 mRNA, and intestinal IgA. As anticipated, LTbetaR blockade significantly decreased PP cells and MAdCAM-1 mRNA, but not intestinal IgA because chow feeding was maintained. Both LTbetaR blockade and PN decreased nasal IgA, but not significantly. CONCLUSIONS: LTbetaR blockade in chow animals significantly reduces transcription of MAdCAM-1 gene and PPs lymphocyte mass. These data implicate inadequate LTbetaR signaling as a major mechanism for decreased GALT cells with lack of enteral stimulation, and further establish the role of LTbetaR in the mucosal immune system.


Subject(s)
Immunity, Mucosal , Immunoglobulin A/immunology , Immunoglobulins/metabolism , Lymphotoxin beta Receptor/antagonists & inhibitors , Mucoproteins/metabolism , Parenteral Nutrition/adverse effects , Peyer's Patches/immunology , Animals , Cell Adhesion Molecules , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation , Humans , Immunity, Mucosal/physiology , Immunoglobulin A/biosynthesis , Intestine, Small/immunology , Lymphocyte Count , Lymphotoxin beta Receptor/metabolism , Male , Mice , Mice, Inbred ICR , Peyer's Patches/cytology , Peyer's Patches/metabolism , Polymerase Chain Reaction/methods , RNA, Messenger/metabolism , Random Allocation
19.
J Neurosci ; 27(28): 7429-37, 2007 Jul 11.
Article in English | MEDLINE | ID: mdl-17626203

ABSTRACT

Inflammation mediated by macrophages is increasingly found to play a central role in diseases and disorders that affect a myriad of organs, prominent among these are diseases of the CNS. The neurotoxicant-induced, cuprizone model of demyelination is ideally suited for the analysis of inflammatory events. Demyelination on exposure to cuprizone is accompanied by predictable microglial activation and astrogliosis, and, after cuprizone withdrawal, this activation reproducibly diminishes during remyelination. This study demonstrates enhanced expression of lymphotoxin beta receptor (Lt betaR) during the demyelination phase of this model, and Lt betaR is found in areas enriched with microglial and astroglial cells. Deletion of the Lt betaR gene (Lt betaR-/-) resulted in a significant delay in demyelination but also a slight delay in remyelination. Inhibition of Lt betaR signaling by an Lt betaR-Ig fusion decoy protein successfully delayed demyelination in wild-type mice. Unexpectedly, this Lt betaR-Ig decoy protein dramatically accelerated the rate of remyelination, even after the maximal pathological disease state had been reached. This strongly indicates the beneficial role of Lt betaR-Ig in the delay of demyelination and the acceleration of remyelination. The discrepancy between remyelination rates in these systems could be attributed to developmental abnormalities in the immune systems of Lt betaR-/- mice. These findings bode well for the use of an inhibitory Lt betaR-Ig as a candidate biological therapy in demyelinating disorders, because it is beneficial during both demyelination and remyelination.


Subject(s)
Demyelinating Diseases/physiopathology , Lymphotoxin beta Receptor/metabolism , Animals , Apoptosis , Brain/metabolism , Brain/pathology , Cell Division , Corpus Callosum/pathology , Cuprizone/pharmacology , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Immunoglobulin G/genetics , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/deficiency , Lymphotoxin beta Receptor/genetics , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Myelin Sheath/drug effects , Myelin Sheath/ultrastructure , Oligodendroglia , Recombinant Fusion Proteins/pharmacology , Time Factors , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
20.
J Immunol ; 178(9): 5659-67, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17442949

ABSTRACT

Stromal cells play a crucial role in the organogenesis of lymphoid tissues. We previously identified VCAM-1(+) stromal cells in cryptopatches (CP) and isolated lymphoid follicles (ILF) in the small intestine of C57BL/6 mice. Nonhemopoietic stromal cell networks in CP and ILF of adult mice also expressed FDC-M1, CD157 (BP-3), and TNF-related activation-induced cytokine (TRANCE). Individual stromal cells were heterogeneous in their expression of these markers, with not all stromal cells expressing the entire set of stromal cell markers. Expression of VCAM-1, FDC-M1, and CD157 on CP stromal cells was absent in alymphoplasia mice deficient in NF-kappaB-inducing kinase (NIK) and NIK knockout mice. Administration of lymphotoxin beta receptor (LTbetaR)-Ig to wild-type mice on day 13 resulted in the absence of CP on day 20; delaying administration of LTbetaR-Ig until day 18 resulted in an 80% decrease in the number of CP on day 22 and diminished expression of VCAM-1, FDC-M1, and CD157 on the remaining CP. In sharp contrast, TRANCE expression by stromal cells was completely independent of NIK and LTbetaR. In addition, expression of TRANCE in ILF was concentrated just beneath the follicle-associated epithelium, a pattern of polarization that was also observed in Peyer's patches. These findings suggest that TRANCE on stromal cells contributes to the differentiation and maintenance of organized lymphoid aggregates in the small intestine.


Subject(s)
Intestine, Small/immunology , Lymphotoxin-alpha/metabolism , Peyer's Patches/cytology , Peyer's Patches/immunology , RANK Ligand/metabolism , ADP-ribosyl Cyclase/analysis , ADP-ribosyl Cyclase/metabolism , Animals , Antigens, CD/analysis , Antigens, CD/metabolism , Cell Differentiation , Cytokines/metabolism , GPI-Linked Proteins , Immunoglobulins/pharmacology , Lymphoid Tissue/cytology , Lymphoid Tissue/growth & development , Lymphoid Tissue/metabolism , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Knockout , Peyer's Patches/growth & development , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RANK Ligand/analysis , Rats , Stromal Cells/chemistry , Stromal Cells/immunology , Vascular Cell Adhesion Molecule-1/analysis , Vascular Cell Adhesion Molecule-1/metabolism , NF-kappaB-Inducing Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...