Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
J Am Soc Nephrol ; 32(8): 1913-1932, 2021 08.
Article in English | MEDLINE | ID: mdl-34155062

ABSTRACT

BACKGROUND: In autosomal dominant polycystic kidney disease (ADPKD), cyst development and enlargement lead to ESKD. Macrophage recruitment and interstitial inflammation promote cyst growth. TWEAK is a TNF superfamily (TNFSF) cytokine that regulates inflammatory responses, cell proliferation, and cell death, and its receptor Fn14 (TNFRSF12a) is expressed in macrophage and nephron epithelia. METHODS: To evaluate the role of the TWEAK signaling pathway in cystic disease, we evaluated Fn14 expression in human and in an orthologous murine model of ADPKD. We also explored the cystic response to TWEAK signaling pathway activation and inhibition by peritoneal injection. RESULTS: Meta-analysis of published animal-model data of cystic disease reveals mRNA upregulation of several components of the TWEAK signaling pathway. We also observed that TWEAK and Fn14 were overexpressed in mouse ADPKD kidney cysts, and TWEAK was significantly high in urine and cystic fluid from patients with ADPKD. TWEAK administration induced cystogenesis and increased cystic growth, worsening the phenotype in a murine ADPKD model. Anti-TWEAK antibodies significantly slowed the progression of ADPKD, preserved renal function, and improved survival. Furthermore, the anti-TWEAK cystogenesis reduction is related to decreased cell proliferation-related MAPK signaling, decreased NF-κB pathway activation, a slight reduction of fibrosis and apoptosis, and an indirect decrease in macrophage recruitment. CONCLUSIONS: This study identifies the TWEAK signaling pathway as a new disease mechanism involved in cystogenesis and cystic growth and may lead to a new therapeutic approach in ADPKD.


Subject(s)
Cytokine TWEAK/metabolism , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , TWEAK Receptor/metabolism , Adult , Animals , Antibodies, Neutralizing/pharmacology , Apoptosis , Cell Proliferation/drug effects , Cysts/metabolism , Cysts/pathology , Cytokine TWEAK/antagonists & inhibitors , Cytokine TWEAK/genetics , Cytokine TWEAK/pharmacology , Disease Models, Animal , Disease Progression , Female , Fibrosis , Gene Expression , Humans , Macrophage Activation/drug effects , Macrophages , Male , Mice , Middle Aged , NF-kappa B/metabolism , Polycystic Kidney, Autosomal Dominant/physiopathology , Signal Transduction , TWEAK Receptor/genetics
2.
Inflamm Res ; 70(5): 553-568, 2021 May.
Article in English | MEDLINE | ID: mdl-33755760

ABSTRACT

BACKGROUND AND AIM: As a proinflammatory cytokine, tumor necrosis factor-like weak inducer of apoptosis (TWEAK) participates in the progression of renal fibrosis by binding to its receptor, fibroblast growth factor-inducible 14 (Fn14). However, the effect of Fn14 inhibition on tubular epithelial cell-mediated tubulointerstitial fibrosis remains unclear. This study aimed to elucidate the role of TWEAK/Fn14 interaction in the development of experimental tubulointerstitial fibrosis as well as the protective effect of Fn14 knockdown on proximal tubular epithelial cells. METHODS: A murine model of unilateral ureteral obstruction was constructed in both wild-type and Fn14-deficient BALB/c mice, followed by observation of the tubulointerstitial pathologies. RESULTS: Fn14 deficiency ameliorated the pathological changes, including inflammatory cell infiltration and cell proliferation, accompanied by reduced production of profibrotic factors and extracellular matrix deposition. In vitro experiments showed that TWEAK dose-dependently enhanced the expression of collagen I, fibronectin, and α-smooth muscle actin in proximal tubular epithelial cells. Interestingly, TWEAK also upregulated the expression levels of Notch1/Jagged1. Fn14 knockdown and Notch1/Jagged1 inhibition also mitigated the effect of TWEAK on these cells. CONCLUSIONS: In conclusion, TWEAK/Fn14 signals contributed to tubulointerstitial fibrosis by acting on proximal tubular epithelial cells. Fn14 inhibition might be a therapeutic strategy for protecting against renal interstitial fibrosis.


Subject(s)
Fibroblast Growth Factors/genetics , Kidney Tubules, Proximal/pathology , Ureteral Obstruction/pathology , Actins/genetics , Actins/metabolism , Animals , Cell Line , Collagen Type I/genetics , Collagen Type I/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Cytokine TWEAK/genetics , Cytokine TWEAK/pharmacology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Fibrosis , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Kidney Tubules, Proximal/metabolism , Male , Mice, Inbred BALB C , Mice, Knockout , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Recombinant Proteins/pharmacology , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism
3.
J Am Soc Nephrol ; 31(9): 2026-2042, 2020 09.
Article in English | MEDLINE | ID: mdl-32631974

ABSTRACT

BACKGROUND: CKD leads to vitamin D deficiency. Treatment with vitamin D receptor agonists (VDRAs) may have nephroprotective and anti-inflammatory actions, but their mechanisms of action are poorly understood. METHODS: Modulation of the noncanonical NF-κB2 pathway and its component TNF receptor-associated factor 3 (TRAF3) by the VDRA paricalcitol was studied in PBMCs from patients with ESKD, cytokine-stimulated cells, and preclinical kidney injury models. RESULTS: In PBMCs isolated from patients with ESKD, TRAF3 protein levels were lower than in healthy controls. This finding was associated with evidence of noncanonical NF-κB2 activation and a proinflammatory state. However, PBMCs from patients with ESKD treated with paricalcitol did not exhibit these features. Experiments in cultured cells confirmed the link between TRAF3 and NF-κB2/inflammation. Decreased TRAF3 ubiquitination in K48-linked chains and cIAP1-TRAF3 interaction mediated the mechanisms of paricalcitol action.TRAF3 overexpression by CRISPR/Cas9 technology mimicked VDRA's effects. In a preclinical model of kidney injury, paricalcitol inhibited renal NF-κB2 activation and decreased renal inflammation. In VDR knockout mice with renal injury, paricalcitol prevented TRAF3 downregulation and NF-κB2-dependent gene upregulation, suggesting a VDR-independent anti-inflammatory effect of paricalcitol. CONCLUSIONS: These data suggest the anti-inflammatory actions of paricalcitol depend on TRAF3 modulation and subsequent inhibition of the noncanonical NF-κB2 pathway, identifying a novel mechanism for VDRA's effects. Circulating TRAF3 levels could be a biomarker of renal damage associated with the inflammatory state.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Ergocalciferols/pharmacology , Kidney Failure, Chronic/drug therapy , Receptors, Calcitriol/agonists , TNF Receptor-Associated Factor 3/physiology , Animals , Cells, Cultured , Cytokine TWEAK/pharmacology , Female , Humans , Male , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Receptors, Calcitriol/physiology , Signal Transduction/drug effects , TNF Receptor-Associated Factor 3/analysis
4.
Nanotechnology ; 30(46): 465101, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31323657

ABSTRACT

In the last ten years, there has been little advancement in the treatment of the aggressive brain cancer Glioblastoma Multiforme (GBM). This research describes the synthesis of a superparamagnetic iron oxide (SPION)-based nanotheraputic complex for use in targeting and killing aggressive mesenchymal GBM cells. The average sizes and magnetic properties of the synthesized SPIONs are tailored via a novel time-controlled approach to a previously described electrochemical reaction. Through this synthetic method, the optimal particle size where maximal thermal energy is released upon stimulation with an external magnetic field was determined to be 21 nm. The nano-complex was further modified to selectively target GBM cells by adding a heterobifunctional poly(ethylene) glycol polymer crosslinked to TWEAK (a GBM targeting peptide). Preliminary investigation with FITC Annexin V/propidium iodide fluorescent probes and transmission electron microscopy revealed biochemical and morphological evidence of both SPION internalization and cytotoxic effects over the course of three hours. Thus, these nano-complexes hold promise as a potential treatment agent for an otherwise untreatable disease.


Subject(s)
Brain Neoplasms/metabolism , Cytokine TWEAK/pharmacology , Ferric Compounds/pharmacology , Glioblastoma/metabolism , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Cross-Linking Reagents , Cytokine TWEAK/chemistry , Ferric Compounds/chemistry , Glioblastoma/drug therapy , Humans , Magnetite Nanoparticles/chemistry , Microscopy, Electron, Transmission , Particle Size , Polyethylene Glycols/chemistry
5.
Exp Lung Res ; 44(7): 332-343, 2018 09.
Article in English | MEDLINE | ID: mdl-30676129

ABSTRACT

AIM OF THE STUDY: In patients with asthma, chronic inflammatory processes and the subsequent remodeling of the airways contribute to the symptoms and the pathophysiological changes. Epithelial-mesenchymal transition (EMT) is thought to play an important role in tissue remodeling. Previous reports show that tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a cytokine of the TNF superfamily, exerts pro-inflammatory effects, and enhances transforming growth factor (TGF)-ß-induced EMT in bronchial epithelial cells. In this study, we investigated the TWEAK-induced cytokine and chemokine production in the human bronchial epithelial cell line BEAS-2B during EMT. MATERIALS AND METHODS: Quantitative real-time RT-PCR, enzyme-linked immunosorbent assays, western blotting, and immunohistochemistry were used to define the production of cytokines and chemokines. RESULTS: We found that TWEAK increases mRNA and protein levels of thymic stromal lymphopoietin (TSLP), monocyte chemoattractant protein -1 (MCP-1), regulated upon activation normal T cell express sequence (RANTES), and IL-8 in BEAS-2B bronchial epithelial cells. Moreover, co-treatment with TWEAK and TGF-ß1 induces not only features of EMT but also enhances the production of TSLP and RANTES. Thymus- and activation-regulated chemokines (TARC) production is induced by the co-treatment of TWEAK and TGF-ß1 but not by TWEAK or TGF-ß1 stimulation alone. Furthermore, the increased mRNA expression of TSLP and RANTES after co-treatment with TWEAK and TGF-ß1 is prevented by inhibitors of Smad-independent signaling pathways. CONCLUSIONS: In the present study, we have revealed a novel mechanism for the production of asthma-related cytokines and chemokines in EMT driven by the co-stimulation with TWEAK and TGF-ß1. We conclude that cellular EMT processes caused by TWEAK and TGF-ß1 may contribute to chronic airway inflammation and remodeling.


Subject(s)
Chemokine CCL17/biosynthesis , Chemokine CCL5/biosynthesis , Cytokine TWEAK/pharmacology , Cytokines/biosynthesis , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Transforming Growth Factor beta1/pharmacology , Airway Remodeling , Asthma/metabolism , Bronchi/cytology , Cell Line , Humans , Thymic Stromal Lymphopoietin
6.
J Biomed Mater Res A ; 105(9): 2551-2561, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28512793

ABSTRACT

Incomplete regeneration after trauma or muscular dysfunction is a common problem in muscle replacement therapies. Recent approaches in tissue engineering allow for the replication of skeletal muscle structure and function in vitro and in vivo by molecular therapies and implantable scaffolds which properly address muscle cells toward myotube differentiation and maturation. Here, we investigate the in vitro response of human mesenchymal stem cells (hMSC) on electrospun fibers made of polycaprolactone (PCL) in the presence of 5-azacytidine (5-AZA) to evaluate how fibrous network may influence the therapeutic effect of drug during in vitro myogenesis. Biological studies demonstrate the ability of hMSCs to differentiate in mature myofibers in supplemented (myogenic) and, preferentially, in 5-AZA-enriched culture. PCL electrospun fibers amplify the 5-AZA capability to induce a low proliferation rate in hMSC, thus promoting hMSC differentiation (MTT assay). Qualitative (Azan Mallory stain, immunofluorescence assay, SEM analyses) and quantitative (ELISA test) assays confirm the synergistic contribution of PCL electrospun fibers and 5-AZA on in vitro myotubes formation and maturation. This result is also confirmed by the expression of muscle-specific proteins related to the myogenic mechanisms in the presence of other muscle inductive signals (i.e., oxytocin, Tweak). Hence, we suggest the use of PCL electrospun fibers as interesting preclinical model to explore the effect of drugs and chemotherapeutics administration after damaged muscle resection. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2551-2561, 2017.


Subject(s)
Azacitidine/pharmacology , Mesenchymal Stem Cells/cytology , Muscle, Skeletal/physiology , Regeneration/drug effects , Tissue Engineering , Tissue Scaffolds/chemistry , Cell Shape/drug effects , Cell Survival/drug effects , Cytokine TWEAK/pharmacology , Humans , Mesenchymal Stem Cells/drug effects , Muscle Development/drug effects , Muscle, Skeletal/drug effects , Oxytocin/pharmacology , Polyesters
7.
Cell Death Differ ; 24(3): 481-491, 2017 03.
Article in English | MEDLINE | ID: mdl-28106882

ABSTRACT

Peptido-mimetic inhibitor of apoptosis protein (IAP) antagonists (Smac mimetics (SMs)) can kill tumour cells by depleting endogenous IAPs and thereby inducing tumour necrosis factor (TNF) production. We found that interferon-γ (IFNγ) synergises with SMs to kill cancer cells independently of TNF- and other cell death receptor signalling pathways. Surprisingly, CRISPR/Cas9 HT29 cells doubly deficient for caspase-8 and the necroptotic pathway mediators RIPK3 or MLKL were still sensitive to IFNγ/SM-induced killing. Triple CRISPR/Cas9-knockout HT29 cells lacking caspase-10 in addition to caspase-8 and RIPK3 or MLKL were resistant to IFNγ/SM killing. Caspase-8 and RIPK1 deficiency was, however, sufficient to protect cells from IFNγ/SM-induced cell death, implying a role for RIPK1 in the activation of caspase-10. These data show that RIPK1 and caspase-10 mediate cell death in HT29 cells when caspase-8-mediated apoptosis and necroptosis are blocked and help to clarify how SMs operate as chemotherapeutic agents.


Subject(s)
Apoptosis/drug effects , Caspase 10/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Interferon-gamma/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , CRISPR-Cas Systems/genetics , Caspase 10/chemistry , Caspase 10/genetics , Caspase 8/chemistry , Caspase 8/genetics , Caspase 8/metabolism , Caspase Inhibitors/pharmacology , Cell Line , Cytokine TWEAK/pharmacology , Drug Synergism , HT29 Cells , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Interferon-gamma/genetics , Interferon-gamma/metabolism , Mice , Mice, Knockout , Pentanoic Acids/pharmacology , Protein Kinases/deficiency , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
8.
Sci Rep ; 6: 28857, 2016 06 29.
Article in English | MEDLINE | ID: mdl-27353019

ABSTRACT

TNF-like weak inducer of apoptosis (TWEAK) receptor Fn14 is expressed by podocytes and Fn14 deficiency protects from experimental proteinuric kidney disease. However, the downstream effectors of TWEAK/Fn14 in podocytes are poorly characterized. We have explored TWEAK activation of non-canonical NFκB signaling in cultured podocytes. In cultured podocytes, TWEAK increased the expression of the chemokines CCL21, CCL19 and RANTES in a time-dependent manner. The inhibitor of canonical NFκB activation parthenolide inhibited the CCL19 and the early RANTES responses, but not the CCL21 or late RANTES responses. In this regard, TWEAK induced non-canonical NFκB activation in podocytes, characterized by NFκB2/p100 processing to NFκB2/p52 and nuclear migration of RelB/p52. Silencing by a specific siRNA of NIK, the upstream kinase of the non-canonical NFκB pathway, prevented CCL21 upregulation but did not modulate CCL19 or RANTES expression in response to TWEAK, thus establishing CCL21 as a non-canonical NFκB target in podocytes. Increased kidney Fn14 and CCL21 expression was also observed in rat proteinuric kidney disease induced by puromycin, and was localized to podocytes. In conclusion, TWEAK activates the non-canonical NFκB pathway in podocytes, leading to upregulation of CCL21 expression. The non-canonical NFκB pathway should be explored as a potential therapeutic target in proteinuric kidney disease.


Subject(s)
Chemokine CCL21/biosynthesis , NF-kappa B/metabolism , Podocytes/metabolism , Signal Transduction , Active Transport, Cell Nucleus , Animals , Cells, Cultured , Chemokine CCL19/genetics , Chemokine CCL19/metabolism , Chemokine CCL21/genetics , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Cytokine TWEAK/pharmacology , Gene Expression , Gene Expression Regulation , Humans , Mice , Protein Processing, Post-Translational , Proteinuria/drug therapy , Rats, Inbred WKY , Renal Insufficiency, Chronic/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL