Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Front Immunol ; 14: 1219907, 2023.
Article in English | MEDLINE | ID: mdl-37465675

ABSTRACT

Fibrosis is the result of extracellular matrix protein deposition and remains a leading cause of death in USA. Despite major advances in recent years, there remains an unmet need to develop therapeutic options that can effectively degrade or reverse fibrosis. The tumor necrosis super family (TNFSF) members, previously studied for their roles in inflammation and cell death, now represent attractive therapeutic targets for fibrotic diseases. In this review, we will summarize select TNFSF and their involvement in fibrosis of the lungs, the heart, the skin, the gastrointestinal tract, the kidney, and the liver. We will emphasize their direct activity on epithelial cells, fibroblasts, and smooth muscle cells. We will further report on major clinical trials targeting these ligands. Whether in isolation or in combination with other anti-TNFSF member or treatment, targeting this superfamily remains key to improve efficacy and selectivity of currently available therapies for fibrosis.


Subject(s)
Epithelial Cells , Kidney , Humans , Fibrosis , Kidney/metabolism , Epithelial Cells/metabolism , Inflammation/metabolism , Lung/metabolism
2.
J Allergy Clin Immunol ; 151(4): 976-990.e5, 2023 04.
Article in English | MEDLINE | ID: mdl-36473503

ABSTRACT

BACKGROUND: Dysregulation of airway smooth muscle cells (ASM) is central to the severity of asthma. Which molecules dominantly control ASM in asthma is unclear. High levels of the cytokine LIGHT (aka TNFSF14) have been linked to asthma severity and lower baseline predicted FEV1 percentage, implying that signals through its receptors might directly control ASM dysfunction. OBJECTIVE: Our study sought to determine whether signaling via lymphotoxin beta receptor (LTßR) or herpesvirus entry mediator from LIGHT dominantly drives ASM hyperreactivity induced by allergen. METHODS: Conditional knockout mice deficient for LTßR or herpesvirus entry mediator in smooth muscle cells were used to determine their role in ASM deregulation and airway hyperresponsiveness (AHR) in vivo. Human ASM were used to study signals induced by LTßR. RESULTS: LTßR was strongly expressed in ASM from normal and asthmatic subjects compared to several other receptors implicated in smooth muscle deregulation. Correspondingly, conditional deletion of LTßR only in smooth muscle cells in smMHCCreLTßRfl/fl mice minimized changes in their numbers and mass as well as AHR induced by house dust mite allergen in a model of severe asthma. Intratracheal LIGHT administration independently induced ASM hypertrophy and AHR in vivo dependent on direct LTßR signals to ASM. LIGHT promoted contractility, hypertrophy, and hyperplasia of human ASM in vitro. Distinguishing LTßR from the receptors for IL-13, TNF, and IL-17, which have also been implicated in smooth muscle dysregulation, LIGHT promoted NF-κB-inducing kinase-dependent noncanonical nuclear factor kappa-light-chain enhancer of activated B cells in ASM in vitro, leading to sustained accumulation of F-actin, phosphorylation of myosin light chain kinase, and contractile activity. CONCLUSIONS: LTßR signals directly and dominantly drive airway smooth muscle hyperresponsiveness relevant for pathogenesis of airway remodeling in severe asthma.


Subject(s)
Asthma , Receptors, Tumor Necrosis Factor, Member 14 , Humans , Mice , Animals , Lymphotoxin beta Receptor/genetics , Asthma/pathology , Muscle, Smooth , Myocytes, Smooth Muscle/pathology , Mice, Knockout , Allergens , Lung/pathology
3.
J Immunol ; 2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36426994

ABSTRACT

Eosinophilic esophagitis (EoE) is a chronic type 2 allergic disease, with esophageal tissue remodeling as the mechanism behind clinical dysphagia and strictures. IL-13 is thought to be a central driver of disease, but other inflammatory factors, such as IFNs and TNF superfamily members, have been hypothesized to play a role in disease pathogenesis. We recently found that the cytokine TNFSF14/LIGHT is upregulated in the esophagus of patients with EoE and that LIGHT promotes inflammatory activity in esophageal fibroblasts. However, the global effects of LIGHT on EoE pathogenesis in vivo remain unknown. We investigated the impact of a LIGHT deficiency in a murine model of EoE driven by house dust mite allergen. Chronic intranasal challenge with house dust mite promoted esophageal eosinophilia and increased CD4+ T cell numbers and IL-13 and CCL11 production in wild-type mice. Esophageal remodeling was reflected by submucosal collagen accumulation, increased muscle density, and greater numbers of fibroblasts. LIGHT-/- mice displayed normal esophageal eosinophilia, but exhibited reduced frequencies of CD4 T cells, IL-13 expression, submucosal collagen, and muscle density and a decrease in esophageal accumulation of fibroblasts. In vitro, LIGHT increased division of human esophageal fibroblasts and selectively enhanced IL-13-mediated expression of a subset of inflammatory and fibrotic genes. These results show that LIGHT contributes to various features of murine EoE, impacting the accumulation of CD4 T cells, IL-13 production, fibroblast proliferation, and esophagus remodeling. These findings suggest that LIGHT may be, to our knowledge, a novel therapeutic target for the treatment of EoE.

4.
J Immunol ; 2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36288906

ABSTRACT

Eosinophilic esophagitis (EoE) is a chronic type 2 allergic disease, with esophageal tissue remodeling as the mechanism behind clinical dysphagia and strictures. IL-13 is thought to be a central driver of disease, but other inflammatory factors, such as IFNs and TNF superfamily members, have been hypothesized to play a role in disease pathogenesis. We recently found that the cytokine TNFSF14/LIGHT is upregulated in the esophagus of patients with EoE and that LIGHT promotes inflammatory activity in esophageal fibroblasts. However, the global effects of LIGHT on EoE pathogenesis in vivo remain unknown. We investigated the impact of a LIGHT deficiency in a murine model of EoE driven by house dust mite allergen. Chronic intranasal challenge with house dust mite promoted esophageal eosinophilia and increased CD4+ T cell numbers and IL-13 and CCL11 production in wild-type mice. Esophageal remodeling was reflected by submucosal collagen accumulation, increased muscle density, and greater numbers of fibroblasts. LIGHT-/- mice displayed normal esophageal eosinophilia, but exhibited reduced frequencies of CD4 T cells, IL-13 expression, submucosal collagen, and muscle density and a decrease in esophageal accumulation of fibroblasts. In vitro, LIGHT increased division of human esophageal fibroblasts and selectively enhanced IL-13-mediated expression of a subset of inflammatory and fibrotic genes. These results show that LIGHT contributes to various features of murine EoE, impacting the accumulation of CD4 T cells, IL-13 production, fibroblast proliferation, and esophagus remodeling. These findings suggest that LIGHT may be, to our knowledge, a novel therapeutic target for the treatment of EoE.

5.
J Immunol ; 208(3): 745-752, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35031577

ABSTRACT

Cystic fibrosis (CF) is an inherited life-threatening disease accompanied by repeated lung infections and multiorgan inflammation that affects tens of thousands of people worldwide. The causative gene, cystic fibrosis transmembrane conductance regulator (CFTR), is mutated in CF patients. CFTR functions in epithelial cells have traditionally been thought to cause the disease symptoms. Recent work has shown an additional defect: monocytes from CF patients show a deficiency in integrin activation and adhesion. Because monocytes play critical roles in controlling infections, defective monocyte function may contribute to CF progression. In this study, we demonstrate that monocytes from CFTRΔF508 mice (CF mice) show defective adhesion under flow. Transplanting CF mice with wild-type (WT) bone marrow after sublethal irradiation replaced most (60-80%) CF monocytes with WT monocytes, significantly improved survival, and reduced inflammation. WT/CF mixed bone marrow chimeras directly demonstrated defective CF monocyte recruitment to the bronchoalveolar lavage and the intestinal lamina propria in vivo. WT mice reconstituted with CF bone marrow also show lethality, suggesting that the CF defect in monocytes is not only necessary but also sufficient to cause disease. We also show that monocyte-specific knockout of CFTR retards weight gains and exacerbates dextran sulfate sodium-induced colitis. Our findings show that providing WT monocytes by bone marrow transfer rescues mortality in CF mice, suggesting that similar approaches may mitigate disease in CF patients.


Subject(s)
Cell Adhesion/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/therapy , Monocytes/immunology , Monocytes/transplantation , Animals , Bone Marrow Transplantation , Bronchoalveolar Lavage Fluid/cytology , Colitis/pathology , Cystic Fibrosis/pathology , Integrins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL
6.
J Immunol Methods ; 497: 113107, 2021 10.
Article in English | MEDLINE | ID: mdl-34352237

ABSTRACT

Understanding the interplay between immune and structural cells is important for studying fibrosis and inflammation; however, primary immune cell isolation from organs that are typically enriched in stromal cells, like the lung, esophagus, or gut, proves to be an ongoing challenge. In fibrotic conditions, this challenge becomes even greater as infiltrating cells become trapped in the robust extracellular matrix (ECM). This protocol details a method to isolate cells at high yield from stroma-rich organs that can be used for further analyses via flow cytometry, stimulation, or culturing. Validation of this method is confirmed by flow cytometry data assessing immune cell populations of interest. This protocol can be completed in approximately 5-6 h.


Subject(s)
Cell Separation , Esophageal Mucosa/cytology , Flow Cytometry , Intestinal Mucosa/cytology , Skin/pathology , Animals , Biomarkers/metabolism , Cell Survival , Cells, Cultured , Collagenases/metabolism , Endopeptidases/metabolism , Esophageal Mucosa/immunology , Esophageal Mucosa/metabolism , Fibrosis , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Skin/immunology , Skin/metabolism , Time Factors , Trypsin/metabolism , Workflow
7.
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
8.
J Immunol ; 205(9): 2414-2422, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32958689

ABSTRACT

Lung fibrosis and tissue remodeling are features of chronic diseases such as severe asthma, idiopathic pulmonary fibrosis, and systemic sclerosis. However, fibrosis-targeted therapies are currently limited. We demonstrate in mouse models of allergen- and bleomycin-driven airway inflammation that neutralization of the TNF family cytokine TL1A through Ab blocking or genetic deletion of its receptor DR3 restricted increases in peribronchial smooth muscle mass and accumulation of lung collagen, primary features of remodeling. TL1A was found as a soluble molecule in the airways and expressed on the surface of alveolar macrophages, dendritic cells, innate lymphoid type 2 cells, and subpopulations of lung structural cells. DR3 was found on CD4 T cells, innate lymphoid type 2 cells, macrophages, fibroblasts, and some epithelial cells. Suggesting in part a direct activity on lung structural cells, administration of recombinant TL1A into the naive mouse airways drove remodeling in the absence of other inflammatory stimuli, innate lymphoid cells, and adaptive immunity. Correspondingly, human lung fibroblasts and bronchial epithelial cells were found to express DR3 and responded to TL1A by proliferating and/or producing fibrotic molecules such as collagen and periostin. Reagents that disrupt the interaction of TL1A with DR3 then have the potential to prevent deregulated tissue cell activity in lung diseases that involve fibrosis and remodeling.


Subject(s)
Airway Remodeling/immunology , Idiopathic Pulmonary Fibrosis/immunology , Lung/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Adaptive Immunity/immunology , Animals , Asthma/immunology , Bleomycin/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Dendritic Cells/immunology , Epithelial Cells/immunology , Female , Humans , Immunity, Innate/immunology , Inflammation/immunology , Lymphocytes/immunology , Macrophages, Alveolar/immunology , Mice , Mice, Inbred C57BL
9.
Gastroenterology ; 159(5): 1778-1792.e13, 2020 11.
Article in English | MEDLINE | ID: mdl-32712105

ABSTRACT

BACKGROUND & AIMS: Eosinophilic esophagitis (EoE) is an antigen-mediated eosinophilic disease of the esophagus that involves fibroblast activation and progression to fibrostenosis. Cytokines produced by T-helper type 2 cells and transforming growth factor beta 1 (TGFß1) contribute to the development of EoE, but other cytokines involved in pathogenesis are unknown. We investigate the effects of tumor necrosis factor superfamily member 14 (TNFSF14, also called LIGHT) on fibroblasts in EoE. METHODS: We analyzed publicly available esophageal CD3+ T-cell single-cell sequencing data for expression of LIGHT. Esophageal tissues were obtained from pediatric patients with EoE or control individuals and analyzed by immunostaining. Human primary esophageal fibroblasts were isolated from esophageal biopsy samples of healthy donors or patients with active EoE. Fibroblasts were cultured; incubated with TGFß1 and/or LIGHT; and analyzed by RNA sequencing, flow cytometry, immunoblots, immunofluorescence, or reverse transcription polymerase chain reaction. Eosinophils were purified from peripheral blood of healthy donors, incubated with interleukin 5, cocultured with fibroblasts, and analyzed by immunohistochemistry. RESULTS: LIGHT was up-regulated in the esophageal tissues from patients with EoE, compared with control individuals, and expressed by several T-cell populations, including T-helper type 2 cells. TNF receptor superfamily member 14 (TNFRSF14, also called HVEM) and lymphotoxin beta receptor are receptors for LIGHT that were expressed by fibroblasts from healthy donors or patients with active EoE. Stimulation of esophageal fibroblasts with LIGHT induced inflammatory gene transcription, whereas stimulation with TGFß1 induced transcription of genes associated with a myofibroblast phenotype. Stimulation of fibroblasts with TGFß1 increased expression of HVEM; subsequent stimulation with LIGHT resulted in their differentiation into cells that express markers of myofibroblasts and inflammatory chemokines and cytokines. Eosinophils tethered to esophageal fibroblasts after LIGHT stimulation via intercellular adhesion molecule-1. CONCLUSIONS: T cells in esophageal tissues from patients with EoE express increased levels of LIGHT compared with control individuals, which induces differentiation of fibroblasts into cells with inflammatory characteristics. TGFß1 increases fibroblast expression of HVEM, a receptor for LIGHT. LIGHT mediates interactions between esophageal fibroblasts and eosinophils via ICAM1. This pathway might be targeted for the treatment of EoE.


Subject(s)
Cell Differentiation , Eosinophilic Esophagitis/metabolism , Esophagus/metabolism , Fibroblasts/metabolism , Inflammation Mediators/metabolism , Paracrine Communication , T-Lymphocytes/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Adolescent , Case-Control Studies , Cells, Cultured , Child , Child, Preschool , Eosinophilic Esophagitis/immunology , Eosinophilic Esophagitis/pathology , Esophagus/immunology , Esophagus/pathology , Female , Fibroblasts/immunology , Fibroblasts/pathology , Humans , Intercellular Adhesion Molecule-1/metabolism , Male , Phenotype , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Signal Transduction , T-Lymphocytes/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Up-Regulation
10.
Nat Commun ; 10(1): 823, 2019 02 18.
Article in English | MEDLINE | ID: mdl-30778076

ABSTRACT

Follicular helper T cells (Tfh) play critical roles instructing, and initiating T-cell dependent antibody responses. The underlying mechanisms that enhance their function is therefore critical for vaccine development. Here we apply gene array analysis identifying adenosine deaminase (ADA) as a key molecule that delineates a human Tfh helper program in proliferating circulating Tfh (cTfh) cells and Germinal Centers Tfh (GC-Tfh). ADA-1 expression and enzymatic activity are increased in efficient cTfh2-17/GC-Tfh cells. Exogenous ADA-1 enhances less efficient cTfh1 and pro-follicular Tfh PD-1+ CXCR5+ cells to provide B cell help, while pharmacological inhibition of ADA-1 activity impedes cTfh2-17/GC-Tfh function and diminished antibody response. Mechanistically, ADA-1 controls the Tfh program by influencing IL6/IL-2 production, controlling CD26 extracellular expression and could balance signals through adenosine receptors. Interestingly, dysfunctional Tfh from HIV infected-individual fail to regulate the ADA pathway. Thus, ADA-1 regulates human Tfh and represents a potential target for development of vaccine strategy.


Subject(s)
Adenosine Deaminase/metabolism , HIV Infections/pathology , T-Lymphocytes, Helper-Inducer/physiology , Adenosine Deaminase/genetics , Adenylyl Cyclases/metabolism , B-Lymphocytes/cytology , Coculture Techniques , Dipeptidyl Peptidase 4/metabolism , Germinal Center/metabolism , HIV Infections/metabolism , Humans , Interleukin-2/metabolism , Interleukin-6/metabolism , T-Lymphocytes, Helper-Inducer/virology
11.
J Exp Med ; 215(2): 415-422, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29339444

ABSTRACT

Dermatitis is often associated with an allergic reaction characterized by excessive type 2 responses leading to epidermal acanthosis, hyperkeratosis, and dermal inflammation. Although factors like IL-4, IL-13, and thymic stromal lymphopoietin (TSLP) are thought to be instrumental for the development of this type of skin disorder, other cytokines may be critical. Here, we show that the tumor necrosis factor (TNF) superfamily protein LIGHT (homologous to lymphotoxin, exhibits inducible expression, and competes with HSV glycoprotein D for binding to HVEM, a receptor expressed on T lymphocytes) is required for experimental atopic dermatitis, and LIGHT directly controls keratinocyte hyperplasia, and production of periostin, a matricellular protein that contributes to the clinical features of atopic dermatitis as well as other skin diseases such as scleroderma. Mice with a conditional deletion of the LIGHT receptor HVEM (herpesvirus entry mediator) in keratinocytes phenocopied LIGHT-deficient mice in exhibiting reduced epidermal thickening and dermal collagen deposition in a model of atopic dermatitis driven by house dust mite allergen. LIGHT signaling through HVEM in human epidermal keratinocytes directly induced proliferation and periostin expression, and both keratinocyte-specific deletion of HVEM or antibody blocking of LIGHT-HVEM interactions after disease onset prevented expression of periostin and limited atopic dermatitis symptoms. Developing reagents that neutralize LIGHT-HVEM signaling might be useful for therapeutic intervention in skin diseases where periostin is a central feature.


Subject(s)
Dermatitis, Atopic/metabolism , Keratinocytes/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Animals , Antigens, Dermatophagoides/adverse effects , Cell Adhesion Molecules/metabolism , Cell Proliferation , Dermatitis, Atopic/etiology , Dermatitis, Atopic/immunology , Disease Models, Animal , HEK293 Cells , Humans , Keratinocytes/immunology , Keratinocytes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Small Interfering/genetics , Receptors, Tumor Necrosis Factor, Member 14/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 14/deficiency , Receptors, Tumor Necrosis Factor, Member 14/genetics , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 14/antagonists & inhibitors , Tumor Necrosis Factor Ligand Superfamily Member 14/deficiency , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
12.
Am J Transplant ; 18(7): 1636-1645, 2018 07.
Article in English | MEDLINE | ID: mdl-29266762

ABSTRACT

Calcineurin inhibitor toxicity (CNT) is a frequent occurrence in transplanted renal grafts and autochthone kidneys from patients undergoing long-term treatment with calcineurin inhibitors, notably cyclosporin A (CsA) and tacrolimus. Here, we show an indispensable role of the tumor necrosis factor superfamily (TNFS) molecule TNF-related weak inducer of apoptosis (TWEAK) (TNFSF12) in the pathogenesis of acute CNT lesions in mice. A deficiency in TWEAK resulted in limited tubulotoxicity after CsA exposure, which correlated with diminished expression of inflammatory cytokines and reduced intraparenchymal infiltration with immune cells. We further identified tubular epithelial cells of the kidney as major targets of CsA activity and found that Fn14 (tumor necrosis factor receptor superfamily 12A), the receptor for TWEAK, is a highly CsA-inducible gene in these cells. Correlating with this, CsA pretreatment sensitized tubular epithelial cells specifically to the pro-inflammatory activities of recombinant TWEAK in vitro. Moreover, injection of rTWEAK alone into mice induced moderate disease similar to CsA, and rTWEAK combined with CsA resulted in synergistic nephrotoxicity. These findings support the importance of tubular epithelial cells as cellular targets of CsA toxicity and introduce TWEAK as a critical contributor to CNT pathogenesis.


Subject(s)
Calcineurin Inhibitors/adverse effects , Cytokine TWEAK/metabolism , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Kidney Tubules/pathology , TWEAK Receptor/metabolism , Animals , Cells, Cultured , Cytokine TWEAK/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Male , Mice , Mice, Inbred C57BL , TWEAK Receptor/genetics
13.
Nat Commun ; 8: 15395, 2017 05 22.
Article in English | MEDLINE | ID: mdl-28530223

ABSTRACT

Atopic dermatitis (AD) and psoriasis are driven by alternate type 2 and type 17 immune responses, but some proteins might be critical to both diseases. Here we show that a deficiency of the TNF superfamily molecule TWEAK (TNFSF12) in mice results in defective maintenance of AD-specific T helper type 2 (Th2) and psoriasis-specific Th17 cells in the skin, and impaired expression of disease-characteristic chemokines and cytokines, such as CCL17 and TSLP in AD, and CCL20 and IL-19 in psoriasis. The TWEAK receptor, Fn14, is upregulated in keratinocytes and dermal fibroblasts, and TWEAK induces these cytokines and chemokines alone and in synergy with the signature T helper cytokines of either disease, IL-13 and IL-17. Furthermore, subcutaneous injection of recombinant TWEAK into naive mice induces cutaneous inflammation with histological and molecular signs of both diseases. TWEAK is therefore a critical contributor to skin inflammation and a possible therapeutic target in AD and psoriasis.


Subject(s)
Cytokine TWEAK/genetics , Cytokine TWEAK/metabolism , Dermatitis, Atopic/metabolism , Gene Expression Regulation , Inflammation/metabolism , Psoriasis/metabolism , Animals , Chemokines/metabolism , Interleukin-13/metabolism , Interleukin-17/metabolism , Keratinocytes/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/metabolism , Skin/metabolism , Skin/pathology , TWEAK Receptor/metabolism
14.
Pharmacol Res ; 104: 151-5, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26748035

ABSTRACT

The TNF Superfamily member LIGHT (TNFSF14) has recently emerged as a potential target for therapeutic interventions aiming to halt tissue fibrosis. In this perspective, we discuss how LIGHT may influence the inflammatory and remodeling steps that characterize fibrosis, relevant for many human diseases presenting with scarring such as asthma, idiopathic pulmonary fibrosis, systemic sclerosis, and atopic dermatitis. LIGHT acts through two receptors in the TNF receptor superfamily, HVEM (TNFRSF14) and LTßR (TNFRSF3), which are broadly expressed on hematopoietic and non-hematopoietic cells. LIGHT can regulate infiltrating T cells, macrophages, and eosinophils, controlling their trafficking or retention in the inflamed tissue, their proliferation, and their ability to produce cytokines that amplify fibrotic processes. More interestingly, LIGHT can act on structural cells, namely epithelial cells, fibroblasts, smooth muscle cells, adipocytes, and endothelial cells. By signaling through either HVEM or LTßR expressed on these cells, LIGHT can contribute to their proliferation and expression of chemokines, growth factors, and metalloproteinases. This will lead to hyperplasia of epithelial cells, fibroblasts, and smooth muscle cells, deposition of extracellular matrix proteins, vascular damage, and further immune alterations that in concert constitute fibrosis. Because of its early expression by T cells, LIGHT may be an initiator of fibrotic diseases, but other sources in the immune system could also signify a role for LIGHT in maintaining or perpetuating fibrotic activity. LIGHT may then be an attractive prognostic marker as well as an appealing target for fibrosis therapies relevant to humans.


Subject(s)
Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Animals , Endothelial Cells/metabolism , Eosinophils/metabolism , Epithelial Cells/metabolism , Fibroblasts/metabolism , Fibrosis , Humans , Macrophages/metabolism , Myocytes, Smooth Muscle/metabolism , T-Lymphocytes/metabolism
15.
J Invest Dermatol ; 135(8): 2109-2118, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25789702

ABSTRACT

Several inflammatory diseases including scleroderma and atopic dermatitis display dermal thickening, epidermal hypertrophy, or excessive accumulation of collagen. Factors that might promote these features are of interest for clinical therapy. We previously reported that LIGHT, a TNF superfamily molecule, mediated collagen deposition in the lungs in response to allergen. We therefore tested whether LIGHT might similarly promote collagen accumulation and features of skin fibrosis. Strikingly, injection of recombinant soluble LIGHT into naive mice, either subcutaneously or systemically, promoted collagen deposition in the skin and dermal and epidermal thickening. This replicated the activity of bleomycin, an antibiotic that has been previously used in models of scleroderma in mice. Moreover skin fibrosis induced by bleomycin was dependent on endogenous LIGHT activity. The action of LIGHT in vivo was mediated via both of its receptors, HVEM and LTßR, and was dependent on the innate cytokine TSLP and TGF-ß. Furthermore, we found that HVEM and LTßR were expressed on human epidermal keratinocytes and that LIGHT could directly promote TSLP expression in these cells. We reveal an unappreciated activity of LIGHT on keratinocytes and suggest that LIGHT may be an important mediator of skin inflammation and fibrosis in diseases such as scleroderma or atopic dermatitis.


Subject(s)
Keratinocytes/physiology , Skin/pathology , Skin/physiopathology , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology , Animals , Bleomycin/pharmacology , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Female , Fibrosis/metabolism , Fibrosis/pathology , Fibrosis/physiopathology , Humans , Immunoglobulins/deficiency , Immunoglobulins/genetics , Immunoglobulins/metabolism , Infant, Newborn , Keratinocytes/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, Tumor Necrosis Factor, Member 14/deficiency , Receptors, Tumor Necrosis Factor, Member 14/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Skin/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/deficiency , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
16.
J Allergy Clin Immunol ; 136(3): 757-68, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25680454

ABSTRACT

BACKGROUND: Pulmonary fibrosis is characterized by excessive accumulation of collagen and α-smooth muscle actin in the lung. The key molecules that promote these phenotypes are of clinical interest. OBJECTIVES: Thymic stromal lymphopoietin (TSLP) has been found at high levels in patients with asthma and idiopathic pulmonary fibrosis, and TSLP has been proposed as a primary driver of lung fibrotic disease. We asked whether tumor necrosis factor superfamily protein 14 (TNFSF14) (aka LIGHT) controls TSLP production to initiate fibrosis. METHODS: Expression of TSLP and initiation of pulmonary fibrosis induced by bleomycin were assessed in mice deficient in LIGHT. The ability of recombinant LIGHT, given intratracheally to naive mice, to promote TSLP and fibrosis was also determined. RESULTS: Genetic deletion of LIGHT abolished lung TSLP expression driven by bleomycin, accompanied by near-complete absence of accumulation of lung collagen and α-smooth muscle actin. Furthermore, recombinant LIGHT administered in vivo induced lung expression of TSLP in the absence of other inflammatory stimuli, and strikingly reproduced the primary features of bleomycin-driven disease in a TSLP-dependent manner. Blockade of LIGHT binding to either of its receptors, herpes virus entry mediator and lymphotoxin beta receptor, inhibited clinical symptoms of pulmonary fibrosis, and correspondingly both receptors were found on human bronchial epithelial cells, a primary source of TSLP. Moreover, LIGHT induced TSLP directly in human bronchial epithelial cells and synergized with IL-13 and TGF-ß in vivo to promote TSLP in the lungs and drive fibrosis. CONCLUSIONS: These results show that LIGHT is a profibrogenic cytokine that may be a key driver of TSLP production during the initiation and development of lung fibrotic disease.


Subject(s)
Cytokines/immunology , Lung/immunology , Pulmonary Fibrosis/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Actins/genetics , Actins/immunology , Animals , Bleomycin , Cell Line , Collagen/genetics , Collagen/immunology , Cytokines/genetics , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Fibroblasts/immunology , Fibroblasts/pathology , Gene Expression Regulation , Herpesviridae/immunology , Humans , Lung/pathology , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/immunology , Mice , Mice, Knockout , Protein Binding , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Receptors, Virus/genetics , Receptors, Virus/immunology , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Thymic Stromal Lymphopoietin
17.
J Clin Invest ; 120(6): 1873-84, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20484813

ABSTRACT

The potent regulatory properties of NKT cells render this subset of lipid-specific T cells a promising target for immunotherapeutic interventions. The marine sponge glycolipid alpha-galactosylceramide (alphaGalCer) is the proto-typic NKT cell agonist, which elicits this function when bound to CD1d. However, our understanding of the in vivo properties of NKT cell agonists and the host factors that control their bioactivity remains very limited. In this report, we isolated the enzyme fatty acid amide hydrolase (FAAH) from mouse serum as an alphaGalCer-binding protein that modulates the induction of key effector functions of NKT cells in vivo. FAAH bound alphaGalCer in vivo and in vitro and was required for the efficient targeting of lipid antigens for CD1d presentation. Immunization of Faah-deficient mice with alphaGalCer resulted in a reduced systemic cytokine production, but enhanced expansion of splenic NKT cells. This distinct NKT response conferred a drastically increased adjuvant effect and strongly promoted protective CTL responses. Thus, our findings identify not only the presence of FAAH in normal mouse serum, but also its critical role in the tuning of immune responses to lipid antigens by orchestrating their transport and targeting for NKT cell activation. Our results suggest that the serum transport of lipid antigens directly shapes the quality of NKT cell responses, which could potentially be modulated in support of novel vaccination strategies.


Subject(s)
Amidohydrolases/immunology , Antigens/immunology , Galactosylceramides/immunology , Glycolipids/immunology , Natural Killer T-Cells/immunology , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/pharmacology , Amidohydrolases/metabolism , Animals , Antigens/metabolism , Galactosylceramides/metabolism , Galactosylceramides/pharmacology , Glycolipids/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
18.
J Clin Invest ; 120(5): 1578-90, 2010 May.
Article in English | MEDLINE | ID: mdl-20407212

ABSTRACT

Genetic susceptibility to autoimmunity is frequently associated with specific MHC alleles. Diabetogenic MHC class II molecules, such as human HLA-DQ8 and mouse I-Ag7, typically have a small, uncharged amino acid residue at position 57 of their beta chain (beta57); this results in the absence of a salt bridge between beta57 and Argalpha76, which is adjacent to the P9 pocket of the peptide-binding groove. However, the influence of Argalpha76 on the selection of the TCR repertoire remains unknown, particularly when the MHC molecule binds a peptide with a neutral amino acid residue at position P9. Here, we have shown that diabetogenic MHC class II molecules bound to a peptide with a neutral P9 residue primarily selected and expanded cells expressing TCRs bearing a negatively charged residue in the first segment of their complementarity determining region 3beta. The crystal structure of one such TCR in complex with I-Ag7 bound to a peptide containing a neutral P9 residue revealed that a network of favorable long-range (greater than 4 A) electrostatic interactions existed among Argalpha76, the neutral P9 residue, and TCR, which supported the substantially increased TCR/peptide-MHC affinity. This network could be modulated or switched to a lower affinity interaction by the introduction of a negative charge at position P9 of the peptide. Our results support the existence of a switch at residue beta57 of the I-Ag7 and HLA-DQ8 class II molecules and potentially link normal thymic TCR selection with abnormal peripheral behavior.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Histocompatibility Antigens Class II/genetics , Receptors, Antigen, T-Cell/metabolism , Animals , Cell Separation , Crystallography, X-Ray/methods , Dimerization , Female , Genetic Complementation Test , Genetic Predisposition to Disease , Histocompatibility Antigens Class II/physiology , Humans , Hybridomas/pathology , Kinetics , Mice , Molecular Conformation , Thymus Gland/metabolism
19.
Biochim Biophys Acta ; 1754(1-2): 118-25, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16182622

ABSTRACT

HPr kinase/phosphorylase phosphorylates HPr, a phosphocarrier protein of the phosphoenolpyruvate:carbohydrate phosphotransferase system, at serine-46. P-Ser-HPr is the central regulator of carbon metabolism in Gram-positive bacteria, but also plays a role in virulence development of certain pathogens. In Listeria monocytogenes, several virulence genes, which depend on the transcription activator PrfA, are repressed by glucose, fructose, etc., in a catabolite repressor (CcpA)-independent mechanism. However, the catabolite co-repressor P-Ser-HPr was found to inhibit the activity of PrfA. In an hprKV267F mutant, in which most of the HPr is transformed into P-Ser-HPr, PrfA was barely active. The ptsH1 mutation (Ser-46 of HPr replaced with an alanine) prevented the inhibitory effect of the hprKV267F mutation. Interestingly, disruption of ccpA also inhibited PrfA activity. This effect is probably also mediated via P-Ser-HPr, since ccpA disruption leads to elevated amounts of P-Ser-HPr. Indeed, a ccpA ptsH1 double mutant exhibited normal PrfA activity. In S. pyogenes, the expression of several virulence genes depends on the transcription activator Mga. Interestingly, the mga promoter is preceded by an operator site, which serves as target for the CcpA/P-Ser-HPr complex. Numerous Gram-negative pathogens also contain hprK, which is often organised in an operon with transcription regulators necessary for the development of virulence, indicating that in these organisms P-Ser-HPr also plays a role in pathogenesis. Indeed, inactivation of Neisseria meningitidis hprK strongly diminished cell adhesion of this pathogen.


Subject(s)
Carbohydrate Metabolism , Gram-Negative Bacteria/pathogenicity , Phosphoprotein Phosphatases/physiology , Virulence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Bacterial Proteins/physiology , Carbohydrate Metabolism/physiology , Gram-Negative Bacteria/enzymology , Gram-Positive Bacteria/genetics , Gram-Positive Bacteria/metabolism , Gram-Positive Bacteria/pathogenicity , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Models, Biological , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/pharmacology , Phosphorylation , Streptococcus pyogenes/genetics , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/pathogenicity
20.
J Mol Microbiol Biotechnol ; 9(3-4): 224-34, 2005.
Article in English | MEDLINE | ID: mdl-16415595

ABSTRACT

Listeria monocytogenes PrfA, a transcription activator for several virulence genes, including the hemolysin-encoding hly, is inhibited by rapidly metabolizable carbon sources (glucose, fructose, etc.). This inhibition is not mediated via the major carbon catabolite repression mechanism of gram-positive bacteria, since inactivation of the catabolite control protein A (CcpA) did not prevent the repression of virulence genes by the above sugars. In order to test whether the catabolite co-repressor P-Ser-HPr might be involved in PrfA regulation, we used a Bacillus subtilis strain (BUG1199) containing L. monocytogenes prfA under control of pspac and the lacZ reporter gene fused to the PrfA-activated hly promoter. Formation of P-Ser-HPr requires the bifunctional HPr kinase/phosphorylase (HprK/P), which, depending on the concentration of certain metabolites, either phosphorylates HPr at Ser-46 or dephosphorylates P-Ser-HPr. The hprKV267F allele codes for an HprK/P leading to the accumulation of P-Ser-HPr, since it has normal kinase, but almost no phosphorylase activity. Interestingly, introducing hprKV267F into BUG1199 strongly inhibited transcription activation by PrfA. Preventing the accumulation of P-Ser-HPr in the hprKV267F mutant by replacing Ser-46 in HPr with an alanine restored PrfA activity, while ccpA inactivation had no effect. Interestingly, disruption of ccpA in the hprK wild-type strain BUG1199 also led to inhibition of PrfA. The lowered lacZ expression in the ccpA strain is probably also due to elevated amounts of P-Ser-HPr, since it disappeared when Ser-46 in HPr was replaced with an alanine. To carry out its catalytic function in sugar transport, HPr of the phosphotransferase system (PTS) is also phosphorylated by phosphoenolpyruvate and enzyme I at His-15. However, P-Ser-HPr is only very slowly phosphorylated by enzyme I, which probably accounts for PrfA inhibition. In agreement with this concept, disruption of the enzyme I- or HPr-encoding genes also strongly inhibited PrfA activity. PrfA activity therefore seems to depend on a fully functional PTS phosphorylation cascade.


Subject(s)
Bacterial Proteins/physiology , Listeria monocytogenes/physiology , Peptide Termination Factors/antagonists & inhibitors , Phosphoenolpyruvate Sugar Phosphotransferase System/physiology , Amino Acid Substitution , Artificial Gene Fusion , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Toxins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Heat-Shock Proteins/genetics , Hemolysin Proteins , Listeria monocytogenes/genetics , Models, Biological , Mutation , Peptide Termination Factors/genetics , Phosphoenolpyruvate Sugar Phosphotransferase System/genetics , Phosphotransferases (Nitrogenous Group Acceptor)/genetics , Phosphotransferases (Nitrogenous Group Acceptor)/physiology , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Repressor Proteins/genetics , Repressor Proteins/physiology , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...