Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Sci Rep ; 9(1): 4521, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30872777

ABSTRACT

Systemic sclerosis (SSc or scleroderma) is an auto-immune disease characterized by skin fibrosis. While primary cells from patients are considered as a unique resource to better understand human disease biology, the effect of in vitro culture on these cells and their evaluation as a platform to identify disease regulators remain poorly characterized. The goal of our studies was to provide insights into the utility of SSc dermal fibroblast primary cells for therapeutic target discovery. The disease phenotypes of freshly isolated and in vitro cultured SSc dermal fibroblasts were characterized using whole transcriptome profiling, alpha smooth muscle actin (ASMA) expression and cell impedance. SSc dermal fibroblasts retained most of the molecular disease phenotype upon in vitro culture for at least four cell culture passages (approximatively 10 cell doublings). We validated an RNA interference high throughput assay that successfully identified genes affecting the myofibroblast phenotype of SSc skin fibroblasts. These genes included MKL1, RHOA and LOXL2 that were previously proposed as therapeutic anti-fibrotic target, and ITGA5, that has been less studied in fibrosis biology and may be a novel potential modifier of SSc fibroblast biology. Together our results demonstrated the value of carefully-phenotyped SSc dermal fibroblasts as a platform for SSc target and drug discovery.


Subject(s)
Fibroblasts/metabolism , Scleroderma, Systemic/pathology , Actins/antagonists & inhibitors , Actins/genetics , Actins/metabolism , Adult , Aged, 80 and over , Case-Control Studies , Cells, Cultured , Female , Fibroblasts/cytology , Humans , Male , Middle Aged , Phenotype , Principal Component Analysis , RNA, Small Interfering/metabolism , Scleroderma, Systemic/metabolism , Severity of Illness Index , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Transcriptome
2.
Genes (Basel) ; 9(12)2018 Nov 29.
Article in English | MEDLINE | ID: mdl-30501089

ABSTRACT

Fibroblasts/myofibroblasts are the key effector cells responsible for excessive extracellular matrix (ECM) deposition and fibrosis progression in both idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc) patient lungs, thus it is critical to understand the transcriptomic and proteomic programs underlying their fibrogenic activity. We conducted the first integrative analysis of the fibrotic programming in these cells at the levels of gene and microRNA (miRNA) expression, as well as deposited ECM protein to gain insights into how fibrotic transcriptional programs culminate in aberrant ECM protein production/deposition. We identified messenger RNA (mRNA), miRNA, and deposited matrisome protein signatures for IPF and SSc fibroblasts obtained from lung transplants using next-generation sequencing and mass spectrometry. SSc and IPF fibroblast transcriptional signatures were remarkably similar, with enrichment of WNT, TGF-ß, and ECM genes. miRNA-seq identified differentially regulated miRNAs, including downregulation of miR-29b-3p, miR-138-5p and miR-146b-5p in disease fibroblasts and transfection of their mimics decreased expression of distinct sets of fibrotic signature genes as assessed using a Nanostring fibrosis panel. Finally, proteomic analyses uncovered a distinct "fibrotic" matrisome profile deposited by IPF and SSc fibroblasts compared to controls that highlights the dysregulated ECM production underlying their fibrogenic activities. Our comprehensive analyses of mRNA, miRNA, and matrisome proteomic profiles in IPF and SSc lung fibroblasts revealed robust fibrotic signatures at both the gene and protein expression levels and identified novel fibrogenesis-associated miRNAs whose aberrant downregulation in disease fibroblasts likely contributes to their fibrotic and ECM gene expression.

3.
J Immunol ; 197(10): 3806-3819, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27815440

ABSTRACT

Despite being one of the most common rheumatologic diseases, there is still no disease-modifying drug for primary Sjögren's syndrome (pSS). Advancing our knowledge of the target tissue has been limited by the low dimensionality of histology techniques and the small size of human salivary gland biopsies. In this study, we took advantage of a molecularly validated mouse model of pSS to characterize tissue-infiltrating CD4+ T cells and their regulation by the lymphotoxin/LIGHT signaling axis. Novel cell subsets were identified by combining highly dimensional flow and mass cytometry with transcriptomic analyses. Pharmacologic modulation of the LTßR signaling pathway was achieved by treating mice with LTßR-Ig, a therapeutic intervention currently being tested in pSS patients (Baminercept trial NCT01552681). Using these approaches, we identified two novel CD4+ T cell subsets characterized by high levels of PD1: Prdm1+ effector regulatory T cells expressing immunoregulatory factors, such as Il10, Areg, Fgl2, and Itgb8, and Il21+ effector conventional T cells expressing a pathogenic transcriptional signature. Mirroring these observations in mice, large numbers of CD4+PD1+ T cells were detected in salivary glands from Sjögren's patients but not in normal salivary glands or kidney biopsies from lupus nephritis patients. Unexpectedly, LTßR-Ig selectively halted the recruitment of PD1- naive, but not PD1+, effector T cells to the target tissue, leaving the cells with pathogenic potential unaffected. Altogether, this study revealed new cellular players in pSS pathogenesis, their transcriptional signatures, and differential dependency on the lymphotoxin/LIGHT signaling axis that help to interpret the negative results of the Baminercept trial and will guide future therapeutic interventions.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-alpha/metabolism , Salivary Glands/immunology , Sjogren's Syndrome/immunology , Sjogren's Syndrome/physiopathology , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Amphiregulin/genetics , Animals , Biopsy , Clinical Trials as Topic , Disease Models, Animal , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Humans , Interleukin-10/genetics , Interleukins/genetics , Kidney/pathology , Lupus Nephritis/immunology , Lymphotoxin-alpha/genetics , Mice , Salivary Glands/pathology , Signal Transduction , Sjogren's Syndrome/therapy , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory
4.
J Biol Chem ; 291(38): 19826-34, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27451394

ABSTRACT

B cell activating factor of the TNF family (BAFF), also known as B lymphocyte stimulator, is a ligand required for the generation and maintenance of B lymphocytes. In this study, the ability of different monoclonal antibodies to recognize, inhibit, or activate mouse BAFF was investigated. One of them, a mouse IgG1 named Sandy-2, prevented the binding of BAFF to all of its receptors, BAFF receptor, transmembrane activator and calcium modulating ligand interactor, and B cell maturation antigen, at a stoichiometric ratio; blocked the activity of mouse BAFF on a variety of cell-based reporter assays; and antagonized the prosurvival action of BAFF on primary mouse B cells in vitro A single administration of Sandy-2 in mice induced B cell depletion within 2 weeks, down to levels close to those observed in BAFF-deficient mice. This depletion could then be maintained with a chronic treatment. Sandy-2 and a previously described rat IgG1 antibody, 5A8, also formed a pair suitable for the sensitive detection of endogenous circulating BAFF by ELISA or using a homogenous assay. Interestingly, 5A8 and Sandy-5 displayed activities opposite to that of Sandy-2 by stimulating recombinant BAFF in vitro and endogenous BAFF in vivo These tools will prove useful for the detection and functional manipulation of endogenous mouse BAFF and provide an alternative to the widely used BAFF receptor-Fc decoy receptor for the specific depletion of BAFF in mice.


Subject(s)
Antibodies/pharmacology , B-Cell Activating Factor/antagonists & inhibitors , B-Lymphocytes/immunology , Immunoglobulin G/pharmacology , Animals , Antibodies/immunology , B-Cell Activating Factor/genetics , B-Cell Activating Factor/immunology , B-Lymphocytes/pathology , Cell Survival/drug effects , Hyperplasia , Immunoglobulin G/immunology , Lymphocyte Depletion/methods , Mice , Mice, Knockout
5.
Clin Immunol ; 169: 69-79, 2016 08.
Article in English | MEDLINE | ID: mdl-27352977

ABSTRACT

B-cell activating factor (BAFF) levels are increased in rheumatoid arthritis, lupus and primary Sjögren's syndrome (pSS). However, BAFF contribution to pathogenesis is not completely understood. In pSS, immune infiltration of the salivary and lacrimal glands leads to xerostomia and xerophtalmia. Glandular B cell hyperactivation, differentiation into germinal center (GC)-like structures and plasma cell accumulation are histopathological hallmarks that were attributed to increased BAFF. Here, we experimentally tested this hypothesis by overexpressing BAFF in a mouse model of pSS. BAFF overexpression enhanced lymphocytic infiltration and MHCII expression on B cells. Increased BAFF also induced B cell differentiation into GC B cells within the autoimmune target tissue. However, even in these conditions, GC B cells only accounted for <1% of glandular B cells, demonstrating that BAFF is not efficiently promoting ectopic GC formation in pSS and warranting further investigation of therapeutics targeting both BAFF and the related TNF-family member APRIL.


Subject(s)
B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Sjogren's Syndrome/immunology , Animals , Autoimmunity/genetics , Autoimmunity/immunology , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Differentiation/genetics , Cells, Cultured , Flow Cytometry , Gene Expression Profiling/methods , Germinal Center/immunology , Germinal Center/metabolism , Immunohistochemistry , Lacrimal Apparatus/immunology , Lacrimal Apparatus/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Oligonucleotide Array Sequence Analysis , Sjogren's Syndrome/genetics , Sjogren's Syndrome/metabolism , Xerophthalmia/genetics , Xerophthalmia/immunology , Xerophthalmia/metabolism , Xerostomia/genetics , Xerostomia/immunology , Xerostomia/metabolism
6.
J Am Soc Nephrol ; 27(12): 3639-3652, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27026366

ABSTRACT

The identification of the cellular origins of myofibroblasts has led to the discovery of novel pathways that potentially drive myofibroblast perpetuation in disease. Here, we further investigated the role of innate immune signaling pathways in this process. In mice, renal injury-induced activation of pericytes, which are myofibroblast precursors attached to endothelial cells, led to upregulated expression of TNF receptor superfamily member 12a, also known as fibroblast growth factor-inducible 14 (Fn14), by these cells. In live rat kidney slices, administration of the Fn14 ligand, TNF-related weak inducer of apoptosis (TWEAK), promoted pericyte-dependent vasoconstriction followed by pericyte detachment from capillaries. In vitro, administration of TWEAK activated and differentiated pericytes into cytokine-producing myofibroblasts, and further activated established myofibroblasts in a manner requiring canonical and noncanonical NF-κB signaling pathways. Deficiency of Fn14 protected mouse kidneys from fibrogenesis, inflammation, and associated vascular instability after in vivo injury, and was associated with loss of NF-κB signaling. In a genetic model of spontaneous CKD, therapeutic delivery of anti-TWEAK blocking antibodies attenuated disease progression, preserved organ function, and increased survival. These results identify the TWEAK-Fn14 signaling pathway as an important factor in myofibroblast perpetuation, fibrogenesis, and chronic disease progression.


Subject(s)
Kidney Diseases/etiology , Kidney/pathology , Myofibroblasts/physiology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction , Tumor Necrosis Factors/physiology , Animals , Cytokine TWEAK , Disease Progression , Fibrosis/etiology , Mice , TWEAK Receptor
7.
PLoS One ; 11(2): e0150192, 2016.
Article in English | MEDLINE | ID: mdl-26901467

ABSTRACT

Myotonic dystrophy type 1 (DM1), the most common form of muscular dystrophy in adults, is caused by toxic RNAs produced from the mutant DM protein kinase (DMPK) gene. DM1 is characterized by progressive muscle wasting and weakness. Therapeutic strategies have mainly focused on targeting the toxic RNA. Previously, we found that fibroblast growth factor-inducible 14 (Fn14), the receptor for TWEAK, is induced in skeletal muscles and hearts of mouse models of RNA toxicity and that blocking TWEAK/Fn14 signaling improves muscle function and histology. Here, we studied the effect of Tweak deficiency in a RNA toxicity mouse model. The genetic deletion of Tweak in these mice significantly reduced muscle damage and improved muscle function. In contrast, administration of TWEAK in the RNA toxicity mice impaired functional outcomes and worsened muscle histopathology. These studies show that signaling via TWEAK is deleterious to muscle in RNA toxicity and support the demonstrated utility of anti-TWEAK therapeutics.


Subject(s)
Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Tumor Necrosis Factors/metabolism , Animals , Cytokine TWEAK , Disease Models, Animal , Mice , Mice, Knockout , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Signal Transduction , Tumor Necrosis Factors/genetics
8.
J Biol Chem ; 290(26): 16330-42, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25953898

ABSTRACT

The closely related TNF family ligands B cell activation factor (BAFF) and a proliferation-inducing ligand (APRIL) serve in the generation and maintenance of mature B-lymphocytes. Both BAFF and APRIL assemble as homotrimers that bind and activate several receptors that they partially share. However, heteromers of BAFF and APRIL that occur in patients with autoimmune diseases are incompletely characterized. The N and C termini of adjacent BAFF or APRIL monomers are spatially close and can be linked to create single-chain homo- or hetero-ligands of defined stoichiometry. Similar to APRIL, heteromers consisting of one BAFF and two APRILs (BAA) bind to the receptors B cell maturation antigen (BCMA), transmembrane activator and CAML interactor (TACI) but not to the BAFF receptor (BAFFR). Heteromers consisting of one APRIL and two BAFF (ABB) bind to TACI and BCMA and weakly to BAFFR in accordance with the analysis of the receptor interaction sites in the crystallographic structure of ABB. Receptor binding correlated with activity in reporter cell line assays specific for BAFFR, TACI, or BCMA. Single-chain BAFF (BBB) and to a lesser extent single-chain ABB, but not APRIL or single-chain BAA, rescued BAFFR-dependent B cell maturation in BAFF-deficient mice. In conclusion, BAFF-APRIL heteromers of different stoichiometries have distinct receptor-binding properties and activities. Based on the observation that heteromers are less active than BAFF, we speculate that their physiological role might be to down-regulate BAFF activity.


Subject(s)
B-Cell Activating Factor/metabolism , B-Cell Maturation Antigen/metabolism , Transmembrane Activator and CAML Interactor Protein/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , B-Cell Activating Factor/chemistry , B-Cell Activating Factor/genetics , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/metabolism , B-Cell Maturation Antigen/genetics , Dimerization , Humans , Ligands , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Binding , Signal Transduction , Transmembrane Activator and CAML Interactor Protein/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/chemistry , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
9.
Hum Mol Genet ; 24(7): 2035-48, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25504044

ABSTRACT

Myotonic dystrophy type 1 (DM1), the most prevalent muscular dystrophy in adults, is characterized by progressive muscle wasting and multi-systemic complications. DM1 is the prototype for disorders caused by RNA toxicity. Currently, no therapies exist. Here, we identify that fibroblast growth factor-inducible 14 (Fn14), a member of the tumor necrosis factor receptor super-family, is induced in skeletal muscles and hearts of mouse models of RNA toxicity and in tissues from DM1 patients, and that its expression correlates with severity of muscle pathology. This is associated with downstream signaling through the NF-κB pathways. In mice with RNA toxicity, genetic deletion of Fn14 results in reduced muscle pathology and better function. Importantly, blocking TWEAK/Fn14 signaling with an anti-TWEAK antibody likewise improves muscle histopathology and functional outcomes in affected mice. These results reveal new avenues for therapeutic development and provide proof of concept for a novel therapeutic target for which clinically available therapy exists to potentially treat muscular dystrophy in DM1.


Subject(s)
Myotonic Dystrophy/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factors/metabolism , Adult , Animals , Antibodies/administration & dosage , Cytokine TWEAK , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , Middle Aged , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction/drug effects , TWEAK Receptor , Tumor Necrosis Factor Inhibitors , Tumor Necrosis Factors/genetics
10.
Biochem Biophys Res Commun ; 446(4): 1219-1224, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24680686

ABSTRACT

Progressive loss of skeletal muscle mass and strength (sarcopenia) is a major clinical problem in the elderly. Recently, proinflammatory cytokine TWEAK and its receptor Fn14 were identified as key mediators of muscle wasting in various catabolic states. However, the role of the TWEAK-Fn14 pathway in pathological changes in skeletal muscle during aging remains unknown. In this study, we demonstrate that the levels of Fn14 are increased in skeletal muscle of 18-month old (aged) mice compared with adult mice. Genetic ablation of Fn14 significantly increased the levels of specific muscle proteins and blunted the age-associated fiber atrophy in mice. While gene expression of two prominent muscle-specific E3 ubiquitin ligases MAFBx and MuRF1 remained comparable, levels of ubiquitinated proteins and the expression of autophagy-related molecule Atg12 were significantly reduced in Fn14-knockout (KO) mice compared with wild-type mice during aging. Ablation of Fn14 significantly diminished the DNA-binding activity of transcription factor nuclear factor-kappa B (NF-κB), gene expression of various inflammatory molecules, and interstitial fibrosis in skeletal muscle of aged mice. Collectively, our study suggests that the TWEAK-Fn14 signaling axis contributes to age-associated muscle atrophy and fibrosis potentially through its local activation of proteolytic systems and inflammatory pathways.


Subject(s)
Aging , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Animals , Collagen/analysis , Collagen/metabolism , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Gene Deletion , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , NF-kappa B/metabolism , Proteolysis , Signal Transduction , TWEAK Receptor
11.
Cytokine Growth Factor Rev ; 25(2): 215-25, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24444596

ABSTRACT

TNF-like weak inducer of apoptosis (TWEAK), a TNF superfamily ligand, and its bona fide receptor, the TNF receptor superfamily member fibroblast growth factor-inducible 14 (Fn14), represent a pivotal axis for shaping both physiological and pathological tissue responses to acute or chronic injury and disease. In recent years significant advances have been made in delineating the prominent role of TWEAK-Fn14 dyad in regulating skeletal muscle mass and metabolism. Also emerging from the broad study of tissue injury in skeletal muscle and other organs is the role of the TWEAK-Fn14 pathway in promoting fibrosis. This review article highlights recent advancements toward understanding how the TWEAK-Fn14 pathway regulates the response to various skeletal muscle insults and, more broadly, engages multiple mechanisms to drive tissue fibrosis.


Subject(s)
Muscle, Skeletal/physiology , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factors/genetics , Animals , Cytokine TWEAK , Fibrosis/pathology , Humans , Mice , Myositis/pathology , Regeneration/physiology , Signal Transduction , TWEAK Receptor
12.
FASEB J ; 28(3): 1398-411, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24327607

ABSTRACT

Skeletal muscle wasting attributed to inactivity has significant adverse functional consequences. Accumulating evidence suggests that peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and TNF-like weak inducer of apoptosis (TWEAK)-Fn14 system are key regulators of skeletal muscle mass in various catabolic states. While the activation of TWEAK-Fn14 signaling causes muscle wasting, PGC-1α preserves muscle mass in several conditions, including functional denervation and aging. However, it remains unknown whether there is any regulatory interaction between PGC-1α and TWEAK-Fn14 system during muscle atrophy. Here we demonstrate that TWEAK significantly reduces the levels of PGC-1α and mitochondrial content (∼50%) in skeletal muscle. Levels of PGC-1α are significantly increased in skeletal muscle of TWEAK-knockout (KO) and Fn14-KO mice compared to wild-type mice on denervation. Transgenic (Tg) overexpression of PGC-1α inhibited progressive muscle wasting in TWEAK-Tg mice. PGC-1α inhibited the TWEAK-induced activation of NF-κB (∼50%) and dramatically reduced (∼90%) the expression of atrogenes such as MAFbx and MuRF1. Intriguingly, muscle-specific overexpression of PGC-1α also prevented the inducible expression of Fn14 in denervated skeletal muscle. Collectively, our study demonstrates that TWEAK induces muscle atrophy through repressing the levels of PGC-1α. Overexpression of PGC-1α not only blocks the TWEAK-induced atrophy program but also diminishes the expression of Fn14 in denervated skeletal muscle.


Subject(s)
Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Receptors, Tumor Necrosis Factor/physiology , Transcription Factors/physiology , Tumor Necrosis Factors/physiology , Animals , Cytokine TWEAK , Electrophoretic Mobility Shift Assay , Mice , Mice, Transgenic , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , TWEAK Receptor
13.
Clin Ther ; 35(8): 1137-49, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23928094

ABSTRACT

BACKGROUND: Persistent upregulation of signaling by cytokine tumor necrosis factor-like weak inducer of apoptosis (TWEAK) through its receptor fibroblast growth factor-inducible molecule-14 (Fn14) promotes chronic inflammation and tissue destruction. OBJECTIVE: The aim of this study was to explore the safety and tolerability of the TWEAK-blocking monoclonal antibody BIIB023 and determine its pharmacokinetics and effects on TWEAK pathway pharmacodynamic markers in rheumatoid arthritis (RA). METHODS: Phase I, first-in-human, 2-part, multicenter, double-blind, dose-escalation study. Patients were randomized to a single dose of BIIB023 (0.03-20 mg/kg) (n = 38) or placebo (n = 15) as an add-on to methotrexate. Three open-label cohorts of RA patients taking background disease-modifying antirheumatic drugs and stable tumor necrosis factor (TNF) inhibitor therapy (n = 12) received a single-dose of BIIB023 of 2, 10, or 20 mg/kg and were assessed over 70 days. RESULTS: The incidence of treatment-emergent adverse events for the BIIB023 monotherapy cohorts and open-label cohorts of BIIB023 as add-on therapy to TNF inhibitors compared with placebo were 47% and 50% versus 33%, respectively. Serum exposure to BIIB023 increased in a dose-dependent manner from 0.03 to 20 mg/kg, but not in direct proportion to dose level. After administration, the time course of BIIB023 serum concentration was multiphasic and showed expedited elimination when levels decreased to < 10 µg/mL. Serum-soluble TWEAK levels were suppressed at all dose levels by 6 hours post-dose and recovered to baseline between days 7 and 28. A trend toward downward modulation of serum biomarkers of inflammatory response was suggested in monocyte chemoattractant protein 1, inducible protein 10, macrophage inflammatory protein 1ß, and tissue inhibitor of metalloproteinase 1 in the BIIB023 group versus placebo. CONCLUSIONS: Single-dose BIIB023 showed a favorable safety and tolerability profile in RA. Suppression of serum-soluble TWEAK for ≤ 28 days was observed and downward trends in serum biomarkers suggested.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Tumor Necrosis Factor Inhibitors , Administration, Intravenous , Adolescent , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Neutralizing , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/adverse effects , Antirheumatic Agents/pharmacokinetics , Broadly Neutralizing Antibodies , Cytokine TWEAK , Double-Blind Method , Drug Administration Schedule , Drug Therapy, Combination , Female , Humans , Inflammation/drug therapy , Methotrexate/pharmacokinetics , Methotrexate/therapeutic use , Middle Aged , Receptors, Tumor Necrosis Factor/metabolism , TWEAK Receptor , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factors/immunology , Tumor Necrosis Factors/metabolism , Young Adult
14.
FEBS J ; 280(8): 1818-29, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23438059

ABSTRACT

TWEAK (TNF homologue with weak apoptosis-inducing activity) and Fn14 (fibroblast growth factor-inducible protein 14) are members of the tumor necrosis factor (TNF) ligand and receptor super-families. Having observed that Xenopus Fn14 cross-reacts with human TWEAK, despite its relatively low sequence homology to human Fn14, we examined the conservation in tertiary fold and binding interfaces between the two species. Our results, combining NMR solution structure determination, binding assays, extensive site-directed mutagenesis and molecular modeling, reveal that, in addition to the known and previously characterized ß-hairpin motif, the helix-loop-helix motif makes an essential contribution to the receptor/ligand binding interface. We further discuss the insight provided by the structural analyses regarding how the cysteine-rich domains of the TNF receptor super-family may have evolved over time. DATABASE: Structural data are available in the Protein Data Bank/BioMagResBank databases under the accession codes 2KMZ, 2KN0 and 2KN1 and 17237, 17247 and 17252. STRUCTURED DIGITAL ABSTRACT: TWEAK binds to hFn14 by surface plasmon resonance (View interaction) xeFn14 binds to TWEAK by enzyme linked immunosorbent assay (View interaction) TWEAK binds to xeFn14 by surface plasmon resonance (View interaction) hFn14 binds to TWEAK by enzyme linked immunosorbent assay (View interaction).


Subject(s)
Receptors, Tumor Necrosis Factor/chemistry , Tumor Necrosis Factors/chemistry , Xenopus Proteins/chemistry , Amino Acid Sequence , Animals , Cytokine TWEAK , Extracellular Space/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , TWEAK Receptor , Xenopus laevis
15.
Basic Res Cardiol ; 108(2): 325, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23325387

ABSTRACT

Pulmonary arterial hypertension (PAH) is a fatal disease for which no cure is yet available. The leading cause of death in PAH is right ventricular (RV) failure. Previously, the TNF receptor superfamily member fibroblast growth factor-inducible molecule 14 (Fn14) has been associated with different fibrotic diseases. However, so far there is no study demonstrating a causal role for endogenous Fn14 signaling in RV or LV heart disease. The purpose of this study was to determine whether global ablation of Fn14 prevents RV fibrosis and remodeling improving heart function. Here, we provide evidence for a causative role of Fn14 in pulmonary artery banding (PAB)-induced RV fibrosis and dysfunction in mice. Fn14 expression was increased in the RV after PAB. Mice lacking Fn14 (Fn14(-/-)) displayed substantially reduced RV fibrosis and dysfunction following PAB compared to wild-type littermates. Cell culture experiments demonstrated that activation of Fn14 induces collagen expression via RhoA-dependent nuclear translocation of myocardin-related transcription factor-A (MRTF-A)/MAL. Furthermore, activation of Fn14 in vitro caused fibroblast proliferation and myofibroblast differentiation, which corresponds to suppression of PAB-induced RV fibrosis in Fn14(-/-) mice. Moreover, our findings suggest that Fn14 expression is regulated by endothelin-1 (ET-1) in cardiac fibroblasts. We conclude that Fn14 is an endogenous key regulator in cardiac fibrosis and suggest this receptor as potential new target for therapeutic interventions in heart failure.


Subject(s)
Hypertrophy, Right Ventricular/prevention & control , Myocardium/pathology , Receptors, Tumor Necrosis Factor/physiology , Ventricular Dysfunction, Right/prevention & control , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Differentiation , Cell Proliferation , Collagen/metabolism , Cytokine TWEAK , Endothelin-1/physiology , Familial Primary Pulmonary Hypertension , Fibrosis/prevention & control , Fluorescent Antibody Technique , Hypertension, Pulmonary/complications , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Immunohistochemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Myofibroblasts , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction , TWEAK Receptor , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/metabolism , Up-Regulation , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/physiopathology
16.
Front Pharmacol ; 4: 159, 2013 Dec 18.
Article in English | MEDLINE | ID: mdl-24391588

ABSTRACT

Genetic studies of human diseases have identified multiple genetic risk loci for various fibrotic diseases. This has provided insights into the myriad of biological pathways potentially involved in disease pathogenesis. These discoveries suggest that alterations in immune responses, barrier function, metabolism and telomerase activity may be implicated in the genetic risks for fibrotic diseases. In addition to genetic disease-risks, the identification of genetic disease-modifiers associated with disease complications, severity or prognosis provides crucial insights into the biological processes implicated in disease progression. Understanding the biological processes driving disease progression may be critical to delineate more effective strategies for therapeutic interventions. This review provides an overview of current knowledge and gaps regarding genetic disease-risks and genetic disease-modifiers in human fibrotic diseases.

17.
Mol Pharmacol ; 82(5): 948-57, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22909796

ABSTRACT

Reactive astrogliosis is beneficial in many aspects; however, it is also detrimental in some pathological states such as the development of lethal brain tumors. It is therefore crucial to understand the mechanisms regulating astrocyte proliferation. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor family, was shown to stimulate astrocyte proliferation in vitro. Herein, we further characterize the mitogenic potential of TWEAK on central nervous system cells. Among these cells, astrocytes express the highest level of TWEAK and Fn14 transcripts, suggesting that they are particularly sensitive to TWEAK stimulation. Using in vitro model systems, we found that TWEAK was as potent as epidermal growth factor (EGF) (a prototypical astrocyte mitogen) in mediating astrocyte proliferation. However, its mitogenic activity was delayed compared with that of EGF, suggesting distinct mechanisms of action. Using cell signaling pathway inhibitors, neutralizing antibodies, and protein assays, we further show that the mitogenic activity of TWEAK on primary astrocytes requires stimulation of the transforming growth factor-α (TGF-α) and of the epidermal growth factor receptor (EGFR) signaling pathway through extracellular signal-regulated kinase and p38 mitogen-activated protein kinase activation. In aggregates, our data demonstrate that TWEAK acts as a potent astrocyte mitogen through the induction of a TGF-α/EGFR signaling pathway. We anticipate that description of such a mechanism may allow novel approaches to human pathologies associated with astrocyte proliferation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Astrocytes/cytology , ErbB Receptors/physiology , Membrane Proteins/metabolism , Transforming Growth Factor alpha/physiology , Tumor Necrosis Factors/metabolism , Animals , Apoptosis Regulatory Proteins/pharmacology , Astrocytes/metabolism , Cell Proliferation , Cytokine TWEAK , Embryo, Mammalian , Enzyme Activation , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Membrane Proteins/pharmacology , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Primary Cell Culture , Rats , Rats, Wistar , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , TWEAK Receptor , Tumor Necrosis Factors/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Immunol Rev ; 244(1): 99-114, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22017434

ABSTRACT

Our immune system performs the vital function of recognizing and eliminating invading pathogens and malignancies. There is an increasing appreciation that the immune system also actively mediates tissue responses under both physiological and pathological conditions, significantly impacting the inflammatory, fibrogenic, and regenerative components. Likewise, there is a growing understanding of how epithelial, endothelial, and other non-hematopoietic tissue cell types actively contribute to the interplay that shapes tissue responses. While much of the molecular basis underlying the immune regulation of tissue responses remains to be delineated, the tumor necrosis factor (TNF) superfamily ligand/receptor pair of TNF-like weak inducer of apoptosis (TWEAK) and fibroblast growth factor-inducible molecule 14 (Fn14) has now emerged as a key piece of this puzzle. In this review, we first discuss how the usually 'dormant' TWEAK/Fn14 pathway becomes activated specifically in injury and disease contexts. We then summarize how TWEAK-mediated Fn14 signaling triggers a wide range of activities in tissue parenchymal and stromal cells as well as progenitor cells. Finally, we review recent experimental evidence that further supports the functional dichotomy of TWEAK/Fn14 activation in physiological versus pathological tissue responses and its potential therapeutic implications. Whereas transient TWEAK/Fn14 activation promotes productive tissue responses after injury, excessive or persistent TWEAK/Fn14 activation drives pathological tissue responses, leading to progressive damage and degeneration.


Subject(s)
Immunity, Innate , Inflammation/immunology , Muscle, Skeletal/immunology , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction/immunology , Stem Cells/immunology , Tumor Necrosis Factors/immunology , Animals , Apoptosis/immunology , Cytokine TWEAK , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Gene Expression/immunology , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Transgenic , Muscle, Skeletal/cytology , Muscle, Skeletal/injuries , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Stem Cells/metabolism , TWEAK Receptor , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/metabolism
19.
Arthritis Res Ther ; 13(2): R51, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21435232

ABSTRACT

INTRODUCTION: TNF-like weak inducer of apoptosis (TWEAK) has been proposed as a mediator of inflammation and bone erosion in rheumatoid arthritis (RA). This study aimed to investigate TWEAK and TWEAK receptor (Fn14) expression in synovial tissue from patients with active and inactive rheumatoid arthritis (RA), osteoarthritis (OA) and normal controls and assess soluble (s)TWEAK levels in the synovial fluids from patients with active RA and OA. Effects of sTWEAK on osteoclasts and osteoblasts were investigated in vitro. METHODS: TWEAK and Fn14 expression were detected in synovial tissues by immunohistochemistry (IHC). Selected tissues were dual labelled with antibodies specific for TWEAK and lineage-selective cell surface markers CD68, Tryptase G, CD22 and CD38. TWEAK mRNA expression was examined in human peripheral blood mononuclear cells (PBMC) sorted on the basis of their expression of CD22. sTWEAK was detected in synovial fluid from OA and RA patients by ELISA. The effect of sTWEAK on PBMC and RAW 264.7 osteoclastogenesis was examined. The effect of sTWEAK on cell surface receptor activator of NF Kappa B Ligand (RANKL) expression by human osteoblasts was determined by flow cytometry. RESULTS: TWEAK and Fn14 expression were significantly higher in synovial tissue from all patient groups compared to the synovial tissue from control subjects (P < 0.05). TWEAK was significantly higher in active compared with inactive RA tissues (P < 0.05). TWEAK expression co-localised with a subset of CD38+ plasma cells and with CD22+ B-lymphocytes in RA tissues. Abundant TWEAK mRNA expression was detected in normal human CD22+ B cells. Higher levels of sTWEAK were observed in synovial fluids isolated from active RA compared with OA patients. sTWEAK did not stimulate osteoclast formation directly from PBMC, however, sTWEAK induced the surface expression of RANKL by human immature, STRO-1+ osteoblasts. CONCLUSIONS: The expression of TWEAK by CD22+ B cells and CD38+ plasma cells in RA synovium represents a novel potential pathogenic pathway. High levels of sTWEAK in active RA synovial fluid and of TWEAK and Fn14 in active RA tissue, together with the effect of TWEAK to induce osteoblastic RANKL expression, is consistent with TWEAK/Fn14 signalling being important in the pathogenesis of inflammation and bone erosion in RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Receptors, Tumor Necrosis Factor/biosynthesis , Tumor Necrosis Factors/biosynthesis , Aged , B-Lymphocytes/metabolism , Cell Separation , Cytokine TWEAK , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Male , Middle Aged , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Osteoblasts/metabolism , Plasma Cells/metabolism , Receptors, Tumor Necrosis Factor/analysis , Synovial Fluid/chemistry , Synovial Fluid/metabolism , TWEAK Receptor , Tumor Necrosis Factors/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...