Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Neuroinflammation ; 20(1): 106, 2023 May 03.
Article in English | MEDLINE | ID: mdl-37138340

ABSTRACT

TNF signaling is an essential regulator of cellular homeostasis. Through its two receptors TNFR1 and TNFR2, soluble versus membrane-bound TNF enable cell death or survival in a variety of cell types. TNF-TNFRs signaling orchestrates important biological functions such as inflammation, neuronal activity as well as tissue de- and regeneration. TNF-TNFRs signaling is a therapeutic target for neurodegenerative diseases such as multiple sclerosis (MS) and Alzheimer's disease (AD), but animal and clinical studies yielded conflicting findings. Here, we ask whether a sequential modulation of TNFR1 and TNFR2 signaling is beneficial in experimental autoimmune encephalomyelitis (EAE), an experimental mouse model that recapitulates inflammatory and demyelinating aspects of MS. To this end, human TNFR1 antagonist and TNFR2 agonist were administered peripherally at different stages of disease development in TNFR-humanized mice. We found that stimulating TNFR2 before onset of symptoms leads to improved response to anti-TNFR1 therapeutic treatment. This sequential treatment was more effective in decreasing paralysis symptoms and demyelination, when compared to single treatments. Interestingly, the frequency of the different immune cell subsets is unaffected by TNFR modulation. Nevertheless, treatment with only a TNFR1 antagonist increases T-cell infiltration in the central nervous system (CNS) and B-cell cuffing at the perivascular sites, whereas a TNFR2 agonist promotes Treg CNS accumulation. Our findings highlight the complicated nature of TNF signaling which requires a timely balance of selective activation and inhibition of TNFRs in order to exert therapeutic effects in the context of CNS autoimmunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Receptors, Tumor Necrosis Factor, Type II , Receptors, Tumor Necrosis Factor, Type I , Animals , Humans , Mice , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Inflammation , Multiple Sclerosis/metabolism , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type II/agonists , Tumor Necrosis Factor-alpha/metabolism
2.
Front Immunol ; 12: 737401, 2021.
Article in English | MEDLINE | ID: mdl-34790194

ABSTRACT

Successful implantation requires the coordinated migration and invasion of trophoblast cells from out of the blastocyst and into the endometrium. This process relies on signals produced by cells in the maternal endometrium. However, the relative contribution of stroma cells remains unclear. The study of human implantation has major technical limitations, therefore the need of in vitro models to elucidate the molecular mechanisms. Using a recently described 3D in vitro models we evaluated the interaction between trophoblasts and human endometrial stroma cells (hESC), we assessed the process of trophoblast migration and invasion in the presence of stroma derived factors. We demonstrate that hESC promotes trophoblast invasion through the generation of an inflammatory environment modulated by TNF-α. We also show the role of stromal derived IL-17 as a promoter of trophoblast migration through the induction of essential genes that confer invasive capacity to cells of the trophectoderm. In conclusion, we describe the characterization of a cellular inflammatory network that may be important for blastocyst implantation. Our findings provide a new insight into the complexity of the implantation process and reveal the importance of inflammation for embryo implantation.


Subject(s)
Cell Movement , Embryo Implantation , Endometrium/drug effects , Interleukin-17/metabolism , Paracrine Communication/drug effects , Stromal Cells/drug effects , Trophoblasts/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cell Adhesion , Cell Differentiation , Cell Line , Endometrium/immunology , Endometrium/metabolism , Female , Humans , Interleukin-17/genetics , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Secretory Pathway , Signal Transduction , Stromal Cells/immunology , Stromal Cells/metabolism , Trophoblasts/immunology
3.
Cell Death Dis ; 12(4): 360, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33824270

ABSTRACT

In the early 1990s, it has been described that LTα and LTß form LTα2ß and LTαß2 heterotrimers, which bind to TNFR1 and LTßR, respectively. Afterwards, the LTαß2-LTßR system has been intensively studied while the LTα2ß-TNFR1 interaction has been ignored to date, presumably due to the fact that at the time of identification of the LTα2ß-TNFR1 interaction one knew already two ligands for TNFR1, namely TNF and LTα. Here, we show that LTα2ß interacts not only with TNFR1 but also with TNFR2. We furthermore demonstrate that membrane-bound LTα2ß (memLTα2ß), despite its asymmetric structure, stimulates TNFR1 and TNFR2 signaling. Not surprising in view of its ability to interact with TNFR2, LTα2ß is inhibited by Etanercept, which is approved for the treatment of rheumatoid arthritis and also inhibits TNF and LTα.


Subject(s)
Lymphotoxin-alpha/metabolism , Receptors, Tumor Necrosis Factor, Type II/agonists , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , Humans , Membranes/metabolism , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type I/drug effects , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/immunology
4.
Am J Physiol Heart Circ Physiol ; 316(6): H1528-H1537, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30925081

ABSTRACT

Tumor necrosis factor-α (TNFα) is a proinflammatory cytokine that is closely linked to the development of cardiovascular disease. TNFα activates NADPH oxidase 1 (Nox1) and reactive oxygen species (ROS), including superoxide (O2·-), production extracellularly is required for subsequent signaling in vascular smooth muscle cells (VSMCs). Apoptosis signal-regulating kinase 1 (ASK1) is a mitogen-activated protein kinase kinase kinase that is activated by oxidation of associated thioredoxin. The role of ASK1 in Nox1-mediated signaling by TNFα is poorly defined. We hypothesized that ASK1 is required for TNFα receptor endocytosis and subsequent inflammatory TNFα signaling. We employed a knockdown strategy to explore the role of ASK1 in TNFα signaling in VSMCs. siRNA targeting ASK1 had no effect on TNFα-induced extracellular O2·- production. However, siASK1 inhibited receptor endocytosis as well as phosphorylation of two endocytosis-related proteins, dynamin1 and caveolin1. Intracellular O2·- production was subsequently reduced, as were other inflammatory signaling steps including NF-κB activation, IL-6 production, inducible nitric oxide synthase and VCAM expression, and VSMC proliferation. Prolonged exposure to TNFα (24 h) increased tumor necrosis factor receptor (TNFR) subtype 1 and 2 expression, and these effects were also attenuated by siASK1. ASK1 coimmunoprecipitated with both Nox1 and the leucine rich repeat containing 8A anion channel, two essential components of the TNFR1 signaling complex. Activation of ASK1 by autophosphorylation at Thr845 occurs following thioredoxin dissociation, and this requires the presence of Nox1. Thus, Nox1 is part of the multiprotein ASK1 signaling complex. In response to TNFα, ASK1 is activated by Nox1-derived oxidants, and this plays a critical role in translating these ROS into a physiologic response in VSMCs. NEW & NOTEWORTHY Apoptosis signal-regulating kinase 1 (ASK1) drives dynamin1 and caveolin1 phosphorylation and TNFα receptor endocytosis. ASK1 modulates TNFα-induced NF-κB activation, survival, and proliferation. ASK1 and NADPH oxidase 1 (Nox1) physically associate in a multiprotein signaling complex. Nox1 is required for TNFα-induced ASK1 activation.


Subject(s)
Endocytosis , MAP Kinase Kinase Kinase 5/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , NADPH Oxidase 1/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Superoxides/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Cells, Cultured , Endocytosis/drug effects , MAP Kinase Kinase Kinase 5/genetics , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NADPH Oxidase 1/genetics , Receptors, Tumor Necrosis Factor, Type I/agonists , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
5.
Cardiovasc Res ; 115(6): 1092-1101, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30358805

ABSTRACT

AIMS: Neuroinflammation is a common feature in renovascular, obesity-related, and angiotensin II mediated hypertension. There is evidence that increased release of the pro-inflammatory cytokine tumour necrosis factor-α (TNF-α) contributes to the development of the hypertension, but the underlying neural mechanisms are unclear. Here, we investigated whether TNF-α stimulates neurons in the area postrema (AP), a circumventricular organ, to elicit sympathetic excitation, and increases in blood pressure (BP). METHODS AND RESULTS: In rats with renovascular hypertension, AP neurons that expressed TNF-α type-1 receptor (TNFR1) remained constantly activated (expressed c-Fos) and injection of TNFR1 neutralizing antibody into the AP returned BP (systolic: ∼151 mmHg) to normotensive levels (systolic: ∼108 mmHg). Nanoinjection of TNF-α (100 pg/50 nL) into the AP of anaesthetized normotensive rats increased BP (∼16 mmHg) and sympathetic nerve activity, predominantly to the heart (∼53%), but also to the kidneys (∼35%). These responses were abolished by prior injection of a TNFR1 neutralizing antibody (1 ng/50 nL) within the same site. TNFR1 were expressed in the somata of neurons activated by TNF-α that were retrogradely labelled from the rostral ventrolateral medulla (RVLM). CONCLUSION: These findings indicate that in renovascular hypertension, blocking TNFR1 receptors in the AP significantly reduces BP, while activation of TNFR1 expressing neurons in the AP by TNF-α increases BP in normotensive rats. This is mediated, in part, by projections to the RVLM and an increase in both cardiac and renal sympathetic nerve activity. These findings support the notion that proinflammatory cytokines and neuroinflammation are important pathological mechanisms in the development and maintenance of hypertension.


Subject(s)
Area Postrema/metabolism , Arterial Pressure , Heart Rate , Heart/innervation , Hypertension, Renovascular/metabolism , Kidney/innervation , Neurons/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Sympathetic Nervous System/physiopathology , Animals , Antibodies, Neutralizing/administration & dosage , Area Postrema/drug effects , Area Postrema/physiopathology , Arterial Pressure/drug effects , Disease Models, Animal , Heart Rate/drug effects , Hypertension, Renovascular/physiopathology , Male , Neurons/drug effects , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Sympathetic Nervous System/drug effects , Tumor Necrosis Factor-alpha/administration & dosage
6.
Nat Cell Biol ; 19(10): 1237-1247, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28920952

ABSTRACT

TNF is a master proinflammatory cytokine whose pathogenic role in inflammatory disorders can, in certain conditions, be attributed to RIPK1 kinase-dependent cell death. Survival, however, is the default response of most cells to TNF stimulation, indicating that cell demise is normally actively repressed and that specific checkpoints must be turned off for cell death to proceed. We identified RIPK1 as a direct substrate of MK2 in the TNFR1 signalling pathway. Phosphorylation of RIPK1 by MK2 limits cytosolic activation of RIPK1 and the subsequent assembly of the death complex that drives RIPK1 kinase-dependent apoptosis and necroptosis. In line with these in vitro findings, MK2 inactivation greatly sensitizes mice to the cytotoxic effects of TNF in an acute model of sterile shock caused by RIPK1-dependent cell death. In conclusion, we identified MK2-mediated RIPK1 phosphorylation as an important molecular mechanism limiting the sensitivity of the cells to the cytotoxic effects of TNF.


Subject(s)
Apoptosis/drug effects , Fibroblasts/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Shock/chemically induced , Tumor Necrosis Factor-alpha/toxicity , Animals , Cell Line , Cytosol/enzymology , Disease Models, Animal , Enzyme Activation , Female , Fibroblasts/enzymology , Fibroblasts/pathology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Necrosis , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type I/metabolism , Serine , Shock/enzymology , Shock/pathology , Shock/prevention & control , Signal Transduction/drug effects , Time Factors
7.
Toxicol Sci ; 156(1): 252-260, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28115640

ABSTRACT

Clonidine, an α2-adrenoreceptor agonist, is an anti-glaucoma drug clinically used in many developing countries, and its abuse might damage the cornea and impair human vision. However, its cytotoxicity and precise mechanisms need to be elucidated. Herein, we investigated the cytotoxicity of clonidine and its underlying mechanisms, using an in vitro model of human corneal epithelial (HCEP) cells and an in vivo model of cat corneas, respectively. HCEP cells were treated with various doses of clonidine for 1-28 h, resulting in abnormal morphology, decline of cell viability and G1 phase arrest in a time- and/or dose-dependent manner. Moreover, clonidine treatment induced elevation of plasma membrane permeability, phosphatidylserine externalization, DNA fragmentation, and apoptotic body formation in HCEP cells. Furthermore, we found that clonidine treatment resulted in activated caspase-2, -3, -8, and -9, disruption of the mitochondrial transmembrane potential, downregulation of Bcl-2, and upregulation of Bad, cytoplasmic cytochrome c and apoptosis inducing factor, suggesting that clonidine-induced apoptosis is triggered through Fas/TNFR1 death receptors and Bcl-2 family proteins-mediated mitochondria-dependent pathways. Finally, our in vivo results displayed that 0.25% clonidine could induce DNA fragmentation of cat corneal epithelial cells. In summary, our findings suggest that clonidine above 1/32 of its clinical therapeutic dosage is cytotoxic to corneal epithelial cells by inducing cell apoptosis both in vitro and in vivo, and its pro-apoptotic effect on HCEP cells is triggered by a Fas/TNFR1 death receptors-mediated, mitochondria-dependent signaling pathway.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/adverse effects , Apoptosis/drug effects , Clonidine/adverse effects , Epithelium, Corneal/drug effects , Receptors, Tumor Necrosis Factor, Type I/agonists , Signal Transduction/drug effects , fas Receptor/metabolism , Adrenergic alpha-2 Receptor Agonists/administration & dosage , Animals , Caspases/chemistry , Caspases/metabolism , Cats , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/pathology , Cell Membrane/ultrastructure , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , Clonidine/administration & dosage , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Epithelium, Corneal/metabolism , Epithelium, Corneal/pathology , G1 Phase/drug effects , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Microscopy, Electron, Transmission , Receptors, Tumor Necrosis Factor, Type I/metabolism
8.
Appl Physiol Nutr Metab ; 41(12): 1295-1302, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27863204

ABSTRACT

The inflammation induced by obesogenic diets is associated with deposition of fat in the liver. On the other hand, anti-inflammatory and immunosuppressive therapies may impact in body fat storage and in liver lipid dynamics. It is important to study specific inflammatory mediators in this context, since their role on hepatic damage is not fully clarified. This study aimed to evaluate the role of interleukin (IL)-18 and tumor necrosis factor (TNF) receptor in liver dysfunction induced by diet. Male C57BL/6 wild-type (WT), IL-18, and TNF receptor 1 knockout mice (IL-18-/- and TNFR1-/-) were divided according to the experimental diets: chow diet or a high-refined carbohydrate-containing diet. Alanine aminotransferase was quantified by colorimetric analysis. Total fat content in the liver was determined by Folch methods. Levels of TNF, IL-6, IL-4, and IL-13 in liver samples were measured by ELISA assay. IL-18 and TNFR knockout mice fed with chow diet showed higher liver triglycerides deposition than WT mice fed with the same diet (WT: 131.9 ± 24.5; IL-18-/-: 239.4 ± 38.12*; TNF-/-: 179.6 ± 50.45*; *P < 0.01). Furthermore, these animals also showed a worse liver histopathological score and lower levels of TNF, IL-6, IL-4, and IL-13 in the liver. Interestingly, treatment with a high-carbohydrate diet did not exacerbate liver damage in IL-18-/- and TNFR1-/- mice. Our data suggest that IL-18 and TNF may be involved on hepatic homeostasis mainly in a context of a healthy diet.


Subject(s)
Interleukin-18/metabolism , Lipid Metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Tumor Necrosis Factor, Type I/agonists , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Collagen/metabolism , Diet, Carbohydrate Loading/adverse effects , Food Handling , Inflammasomes/metabolism , Interleukin-13/metabolism , Interleukin-18/genetics , Interleukin-4/metabolism , Interleukin-6/metabolism , Liver/immunology , Liver/pathology , Liver/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/pathology , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Triglycerides/metabolism
9.
Basic Clin Pharmacol Toxicol ; 119(4): 405-11, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27097917

ABSTRACT

Oxymatrine (OMT) is a traditional Chinese medicine monomer and has been used for the treatment of chronic viral hepatitis and many other diseases. We aimed to investigate whether OMT could induce hepatotoxicity in mice and explored the preliminary mechanisms of toxic effects. Twenty-four Institute for Cancer Research male mice were randomly divided into four groups: control group, 40, 160 and 320 mg/kg OMT-treated group. OMT was orally administered once daily for 7 days. The OMT-treated group exhibited an improved liver index and increase in serum alanine aminotransferase, aspartate aminotransferase and alkaline phosphatase,augmented liver histological injury, elevated levels of malondialdehyde and tumour necrosis factor alpha (TNF-α) accompanied by the activation of caspase-9/-8/-3, up-regulated expressions of tumour necrosis factor receptor l (TNFR1), TNF receptor-associated structure domain (TRADD) and phosphorylation of stress-activated protein kinase/c-jun N-terminal protein kinases (p-SAPK/JNK). Altogether, these results suggest that OMT at a dose of 320 mg/kg leads to liver damage and is related to the activation of JNK signalling pathway mediated by TNF-α in the liver of mice.


Subject(s)
Alkaloids/adverse effects , Anti-Arrhythmia Agents/adverse effects , Antiviral Agents/adverse effects , Chemical and Drug Induced Liver Injury/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/drug effects , MAP Kinase Signaling System/drug effects , Quinolizines/adverse effects , Alkaloids/administration & dosage , Animals , Anti-Arrhythmia Agents/administration & dosage , Antiviral Agents/administration & dosage , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/physiopathology , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , JNK Mitogen-Activated Protein Kinases/chemistry , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Liver/physiopathology , Male , Mice, Inbred ICR , Oxidative Stress/drug effects , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Quinolizines/administration & dosage , Random Allocation , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , TNF Receptor-Associated Death Domain Protein/agonists , TNF Receptor-Associated Death Domain Protein/genetics , TNF Receptor-Associated Death Domain Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Respir Res ; 16: 144, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26619903

ABSTRACT

BACKGROUND: Inflammatory respiratory diseases are amongst major global health challenges. Lung fibroblasts have been shown to play a key role in lung inflammatory responses. However, their exact role in initiation and maintenance of lung diseases has remained elusive partly due to the limited availability of physiologically relevant in vitro models. Therefore, developing new tools that enable investigating the molecular pathways (e.g. nuclear factor-kappa B (NF-κB) activation) that underpin inflammatory responses in fibroblasts could be a valuable resource for scientists working in this area of research. RESULTS: In order to investigate NF-κB activation in response to pro-inflammatory stimuli in real-time, we first developed two detection systems based on nuclear localization of NF-κB by immunostaining and luciferase reporter assay system. Furthermore using electrospun porous scaffolds, with similar geometry to human lung extracellular matrix, we developed 3D cultures of lung fibroblasts allowing comparing NF-κB activation in response to pro-inflammatory stimuli (i.e. TNF-α) in 2D and 3D. Our data clearly show that the magnitude of NF-κB activation in 2D cultures is substantially higher than 3D cultures. However, unlike 2D cultures, cells in the 3D model remained responsive to TNF-α at higher concentrations. The more subdued and wider dynamic range of NF-κB responses in 3D culture system was associated with a different expression pattern for TNF receptor I in 3D versus 2D cultures collectively reflecting a more in vivo like TNF receptor I expression and NF-κB activation pattern in the 3D system. CONCLUSION: Our data suggest that lung fibroblasts are actively involved in the pathogenesis of lung inflammation by activation of NF-κB signaling pathway. The 3D culture detection system provides a sensitive and biologically relevant tool for investigating different pro-inflammatory events involving lung fibroblasts.


Subject(s)
Cell Culture Techniques , Fibroblasts/metabolism , Inflammation Mediators/metabolism , Lung/metabolism , Pneumonia/metabolism , Signal Transduction , Transcription Factor RelA/metabolism , Cell Line , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/pathology , Genes, Reporter , Humans , Lung/drug effects , Lung/pathology , Pneumonia/pathology , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Time Factors , Transcription Factor RelA/genetics , Transfection , Tumor Necrosis Factor-alpha/pharmacology
11.
Mediators Inflamm ; 2015: 124942, 2015.
Article in English | MEDLINE | ID: mdl-26504355

ABSTRACT

Tumor necrosis factor- (TNF-) α is a proinflammatory cytokine involved in the development and maintenance of inflammatory and neuropathic pain. Its effects are mediated by two receptors, TNF receptor-1 (TNFR-1) and TNF receptor-2 (TNFR-2). These receptors play a crucial role in the sensitization of voltage-gated sodium channels (VGSCs), a key mechanism in the pathogenesis of chronic pain. Using the whole-cell patch-clamp technique, we examined the influence of TNFR-1 and TNFR-2 on VGSCs and TTX-resistant NaV1.8 channels in isolated rat dorsal root ganglion neurons by using selective TNFR agonists. The TNFR-1 agonist R32W (10 pg/mL) caused an increase in the VGSC current (I(Na(V))) by 27.2 ± 5.1%, while the TNFR-2 agonist D145 (10 pg/mL) increased the current by 44.9 ± 2.6%. This effect was dose dependent. Treating isolated NaV1.8 with R32W (100 pg/mL) resulted in an increase in I(NaV(1.8)) by 18.9 ± 1.6%, while treatment with D145 (100 pg/mL) increased the current by 14.5 ± 3.7%. Based on the current-voltage relationship, 10 pg of R32W or D145 led to an increase in I(Na(V)) in a bell-shaped, voltage-dependent manner with a maximum effect at -30 mV. The effects of TNFR activation on VGSCs promote excitation in primary afferent neurons and this might explain the sensitization mechanisms associated with neuropathic and inflammatory pain.


Subject(s)
Ganglia, Spinal/metabolism , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Neurons/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/chemistry , Voltage-Gated Sodium Channels/metabolism , Animals , Cytokines/metabolism , Dose-Response Relationship, Drug , Inflammation , Male , Neuralgia/drug therapy , Neurons, Afferent/drug effects , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type II/agonists
12.
PLoS One ; 7(7): e41238, 2012.
Article in English | MEDLINE | ID: mdl-22848449

ABSTRACT

TNF can trigger two opposing responses: cell survival and cell death. TNFR1 activates caspases that orchestrate apoptosis but some cell types switch to a necrotic death when treated with caspase inhibitors. Several genes that are required to orchestrate cell death by programmed necrosis have been identified, such as the kinase RIP1, but very little is known about the inhibitory signals that keep this necrotic cell death pathway in check. We demonstrate that T cells lacking the regulatory subunit of IKK, NFκB essential modifier (NEMO), are hypersensitive to programmed necrosis when stimulated with TNF in the presence of caspase inhibitors. Surprisingly, this pro-survival activity of NEMO is independent of NFκB-mediated gene transcription. Instead, NEMO inhibits necrosis by binding to ubiquitinated RIP1 to restrain RIP1 from engaging the necrotic death pathway. In the absence of NEMO, or if ubiquitination of RIP1 is blocked, necrosis ensues when caspases are blocked. These results indicate that recruitment of NEMO to ubiquitinated RIP1 is a key step in the TNFR1 signaling pathway that determines whether RIP1 triggers a necrotic death response.


Subject(s)
I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Ubiquitination , Humans , I-kappa B Kinase/genetics , Jurkat Cells , NF-kappa B/genetics , Necrosis/genetics , Necrosis/metabolism , Nuclear Pore Complex Proteins/genetics , RNA-Binding Proteins/genetics , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , T-Lymphocytes/pathology , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Tumor Necrosis Factor-alpha/pharmacology
13.
Toxicol Appl Pharmacol ; 262(3): 247-54, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22626855

ABSTRACT

Our previous study has shown that genistein enhances apoptosis in A549 lung cancer cells induced by trichostatin A (TSA). The precise molecular mechanism underlying the effect of genistein, however, remains unclear. In the present study, we investigated whether genistein enhances the anti-cancer effect of TSA through up-regulation of TNF receptor-1 (TNFR-1) death receptor signaling. We incubated A549 cells with TSA (50 ng/mL) alone or in combination with genistein and then determined the mRNA and protein expression of TNFR-1 as well as the activation of downstream caspases. Genistein at 5 and 10 µM significantly enhanced the TSA-induced decrease in cell number and apoptosis in a dose-dependent manner. The combined treatment significantly increased mRNA and protein expression of TNFR-1 at 6 and 12h, respectively, compared with that of the control group; while TSA alone had no effect. TSA in combination with 10 µM of genistein increased TNFR-1 mRNA and protein expression by about 70% and 40%, respectively. The underlying mechanism for this effect of genistein may be partly associated with the estrogen receptor pathway. The combined treatment also increased the activation of caspase-3 and -10 as well as p53 protein expression in A549 cells. The enhancing effects of genistein on the TSA-induced decrease in cell number and on the expression of caspase-3 in A549 cells were suppressed by silencing TNFR-1 expression. These data demonstrated that the upregulation of TNFR-1 death receptor signaling plays an important role, at least in part, in the enhancing effect of genistein on TSA-induced apoptosis in A549 cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Genistein/pharmacology , Hydroxamic Acids/pharmacology , Lung Neoplasms/drug therapy , Receptors, Tumor Necrosis Factor, Type I/agonists , Apoptosis/drug effects , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Caspases/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Humans , Lung Neoplasms/metabolism , RNA, Small Interfering/metabolism , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects
14.
FASEB J ; 26(7): 2811-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22441986

ABSTRACT

Alzheimer's disease (AD) is associated with an altered immune response, resulting in chronic increased inflammatory cytokine production with a prominent role of TNF-α. TNF-α signals are mediated by two receptors: TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2). Signaling through TNFR2 is associated with neuroprotection, whereas signaling through TNFR1 is generally proinflammatory and proapoptotic. Here, we have identified a TNF-α-induced proinflammatory agent, lipocalin 2 (Lcn2) via gene array in murine primary cortical neurons. Further investigation showed that Lcn2 protein production and secretion were activated solely upon TNFR1 stimulation when primary murine neurons, astrocytes, and microglia were treated with TNFR1 and TNFR2 agonistic antibodies. Lcn2 was found to be significantly decreased in CSF of human patients with mild cognitive impairment and AD and increased in brain regions associated with AD pathology in human postmortem brain tissue. Mechanistic studies in cultures of primary cortical neurons showed that Lcn2 sensitizes nerve cells to ß-amyloid toxicity. Moreover, Lcn2 silences a TNFR2-mediated protective neuronal signaling cascade in neurons, pivotal for TNF-α-mediated neuroprotection. The present study introduces Lcn2 as a molecular actor in neuroinflammation in early clinical stages of AD.


Subject(s)
Acute-Phase Proteins/metabolism , Alzheimer Disease/metabolism , Lipocalins/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Acute-Phase Proteins/cerebrospinal fluid , Acute-Phase Proteins/genetics , Aged , Aged, 80 and over , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Amyloid beta-Peptides/toxicity , Animals , Base Sequence , Brain/metabolism , Brain/pathology , Case-Control Studies , Cells, Cultured , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Female , Gene Expression/drug effects , Glutamic Acid/toxicity , Humans , Inflammation Mediators/cerebrospinal fluid , Inflammation Mediators/metabolism , Lipocalin-2 , Lipocalins/cerebrospinal fluid , Lipocalins/genetics , Male , Mice , Models, Neurological , Neurons/drug effects , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Oncogene Proteins/genetics , Proto-Oncogene Proteins/cerebrospinal fluid , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type II/agonists , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
15.
J Bone Miner Metab ; 29(6): 671-81, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21626455

ABSTRACT

Tumor necrosis factor (TNF)-α exerts its biological function via TNF type 1 and type 2 receptors (TNFR1 and TNFR2). We have previously reported that bone resorption induced by lipopolysaccharide (LPS) in TNFR2-deficient mice is accelerated compared to that in wild-type (WT) mice. Although these results suggested that TNFR2 might have a protective role in bone resorption, we could not exclude the possibility that TNFR2 has no role in bone resorption. To clarify the role of TNFR2, we developed a TNF-α-induced bone resorption model using cholesterol-bearing pullulan nanogel as a TNF-α carrier to minimize the influence of inflammatory cytokines other than TNF-α. Injections of human TNF-α (hTNF), an agonist of mouse TNFR1, stimulated bone resorption lacunae on the calvariae in WT mice, but mouse TNF-α (mTNF), an agonist of both mouse TNFR1 and TNFR2, could not. To eliminate the possibility that the TNFR1 agonistic effects of hTNF were stronger than those of mTNF, we used the same model in TNFR2-deficient mice. Injection of mTNF resulted in clear bone resorption lacunae to the same extent observed after using hTNF in the TNFR2-deficient mice. Histomorphometric analysis of osteoclast number supported the observed changes in bone resorption lacunae. These data suggest that TNFR2 has a protective role in TNF-α-induced bone resorption.


Subject(s)
Bone Resorption/chemically induced , Bone Resorption/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Skull/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Bone Density/drug effects , Bone Density/genetics , Glucans/chemistry , Humans , Mice , Mice, Mutant Strains , Nanogels , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/genetics , Reverse Transcriptase Polymerase Chain Reaction , Skull/cytology , Skull/metabolism
16.
Biochem Biophys Res Commun ; 388(4): 667-71, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19682974

ABSTRACT

Tumor necrosis factor (TNF) plays important roles in host defense and in preventing tumor formation by acting via its receptors, TNFR1 and TNFR2, functions of which are less understood. To this end, we have been isolating TNF receptor-selective mutants using phage display technique. However, generation of a phage library with large repertoire (>10(8)) is impeded by the limited transformation efficiency of Escherichia coli. Therefore, it is currently difficult to create a mutant library containing amino acid substitutions in more than seven residues. To overcome this problem, here we have used two different TNF mutant libraries, each containing random substitutions at six selected amino acid residues, and utilized a gene shuffling method to construct a randomized mutant library containing substitutions at 12 different amino acid residues of TNF. Consequently, using this library, we identified TNF mutants with greater receptor-selectivity and enhanced receptor-specific bioactivity than the existing mutants.


Subject(s)
Protein Engineering/methods , Receptors, Tumor Necrosis Factor, Type II/agonists , Receptors, Tumor Necrosis Factor, Type I/agonists , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Sequence , Cell Line , DNA Shuffling , Humans , Molecular Sequence Data , Mutation , Peptide Library , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
17.
Cell Biol Int ; 33(9): 1026-31, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19524691

ABSTRACT

Treatment of P388D1, a macrophage-like cell line, with staurosporine triggered apoptosis through the activation of caspase-9 and caspase-3. Unexpected effects of staurosporine on the induction of apoptosis were the activation of caspase-8, and an increase of the levels of TNF-alpha. The increased TNF-alpha levels led to activation of caspase-8 by an autocrine effect via the TNF receptor expressed by the P388D1 macrophages. In contrast, P388D1 macrophages that either had been exposed to UV light or treated with dexamethasone did not undergo apoptosis.


Subject(s)
Apoptosis , Macrophages/drug effects , Protein Kinase Inhibitors/pharmacology , Staurosporine/pharmacology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Caspase 3/drug effects , Caspase 3/metabolism , Caspase 8/drug effects , Caspase 8/metabolism , Caspase 9/drug effects , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , DNA Fragmentation/drug effects , Dexamethasone/pharmacology , Macrophages/radiation effects , Mice , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/drug effects , Ultraviolet Rays/adverse effects
18.
Atherosclerosis ; 196(2): 558-64, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17673218

ABSTRACT

OBJECTIVE: TNFalpha is a pro-inflammatory cytokine that induces endothelial dysfunction and promotes atherosclerosis progression. Down-regulation of lysyl oxidase (LOX), a key enzyme in extracellular matrix maturation, by pro-atherogenic risk factors such as LDL and homocysteine, is associated with an impairment of endothelial barrier function. Our hypothesis is that the inflammatory cytokine TNFalpha could also modulate LOX expression/function in endothelial cells. METHODS: The study was carried out in human umbilical vein endothelial cells (HUVEC), porcine aortic endothelial cells (PAEC) and bovine aortic endothelial cells (BAEC). LOX mRNA levels were analysed by real-time PCR and LOX activity was assessed by a high sensitive fluorescent assay. Promoter activity was determined by transient transfection using a luciferase reporter system. RESULTS: TNFalpha decreases LOX mRNA levels in endothelial cells in a dose- and time-dependent manner. The effect of TNFalpha was observed at low concentrations (0.1-1 ng/mL) and was maximal at 2.5 ng/mL (after 21 h). In transfection assays, TNFalpha reduced LOX transcriptional activity to a similar extent than LOX mRNA. Furthermore, TNFalpha decreases endothelial LOX enzymatic activity. By using both TNF receptor (TNFR) agonist and blocking antibodies we determined the involvement of TNFR2 on LOX down-regulation. Moreover, while TNFR-associated factor-2 (TRAF-2) did not mediate signalling events leading to LOX inhibition, PKC inhibitors counteracted the TNFalpha-induced decrease of LOX mRNA levels. Finally, TNFalpha administration significantly reduced vascular LOX expression in rat aorta. CONCLUSIONS: Endothelial dysfunction induced by TNFalpha is associated with a decrease of LOX expression/activity. Thus, LOX seems to be involved in the impairment of endothelial function triggered by different pathological conditions.


Subject(s)
Endothelium, Vascular/physiopathology , Protein-Lysine 6-Oxidase/biosynthesis , Tumor Necrosis Factor-alpha/physiology , Animals , Aorta/cytology , Apoptosis/drug effects , Cattle , Cells, Cultured , Down-Regulation , Endothelium, Vascular/cytology , Humans , Male , Protein Kinase C/physiology , Rats , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type I/physiology , Receptors, Tumor Necrosis Factor, Type II/agonists , Receptors, Tumor Necrosis Factor, Type II/physiology , Sus scrofa , Umbilical Veins/cytology
19.
Biochim Biophys Acta ; 1774(8): 1029-35, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17561456

ABSTRACT

We have previously produced two bioactive lysine-deficient mutants of TNF-alpha (mutTNF-K90R,-K90P) and found that these mutants have bioactivity superior to wild-type TNF (wtTNF). Because these mutants contained same amino acid except for amino acid 90, it is unclear which amino acid residue is optimal for showing bioactivity. We speculated that this amino acid position was exchangeable, and this amino acid substitution enabled the creation of lysine-deficient mutants with enhanced bioactivity. Therefore, we produced mutTNF-K90R variants (mutTNF-R90X), in which R90 was replaced with other amino acids, to assay their bioactivities and investigated the importance of amino acid position 90. As a result, mutTNF-R90X that replaced R90 with lysine, arginine and proline were bioactive, while other mutants were not bioactive. Moreover, these three mutants showed bioactivity as good as or better than wtTNF. R90 replaced with lysine or arginine had especially superior binding affinities. These results suggest that the amino acid position 90 in TNF-alpha is important for TNF-alpha bioactivity and could be altered to improve its bioactivity to generate a "super-agonist".


Subject(s)
Amino Acid Substitution , Mutation, Missense , Receptors, Tumor Necrosis Factor, Type II/agonists , Receptors, Tumor Necrosis Factor, Type I/agonists , Tumor Necrosis Factor-alpha/immunology , Humans , Protein Binding/genetics , Protein Binding/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , Tumor Necrosis Factor-alpha/genetics
20.
J Biol Chem ; 282(27): 19302-12, 2007 Jul 06.
Article in English | MEDLINE | ID: mdl-17500068

ABSTRACT

Tumor necrosis factor alpha (TNFalpha) is a cytokine secreted by macrophages and adipocytes that contributes to the low grade inflammation and insulin resistance observed in obesity. TNFalpha signaling decreases peroxisome proliferator-activated receptor gamma and glucose transporter isoform 4 (GLUT4) expression in adipocytes, impairing insulin action, and this is mediated in part by the yeast Ste20 protein kinase ortholog Map4k4. Here we show that Map4k4 expression is selectively up-regulated by TNFalpha, whereas the expression of the protein kinases JNK1/2, ERK1/2, p38 stress-activated protein kinase, and mitogen-activated protein kinase kinases 4/7 shows little or no response. Furthermore, the cytokines interleukin 1beta (IL-1beta) and IL-6 as well as lipopolysaccharide fail to increase Map4k4 mRNA levels in cultured adipocytes under conditions where TNFalpha elicits a 3-fold effect. Using agonistic and antagonistic antibodies and small interfering RNA (siRNA) against TNFalpha receptor 1 (TNFR1) and TNFalpha receptor 2 (TNFR2), we show that TNFR1, but not TNFR2, mediates the increase in Map4k4 expression. TNFR1, but not TNFR2, also mediates a potent effect of TNFalpha on the phosphorylation of JNK1/2 and p38 stress-activated protein kinase and their downstream transcription factor substrates c-Jun and activating transcription factor 2 (ATF2). siRNA-based depletion of c-Jun and ATF2 attenuated TNFalpha action on Map4k4 mRNA expression. Consistent with this concept, the phosphorylation of ATF2 along with the expression and phosphorylation of c-Jun by TNFalpha signaling was more robust and prolonged compared with that of IL-1beta, which failed to modulate Map4k4. These data reveal that TNFalpha selectively stimulates the expression of a key component of its own signaling pathway, Map4k4, through a TNFR1-dependent mechanism that targets the transcription factors c-Jun and ATF2.


Subject(s)
Activating Transcription Factor 2/metabolism , Mitogen-Activated Protein Kinases/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , 3T3 Cells , Adipocytes/metabolism , Animals , Glucose Transporter Type 4/metabolism , Inflammation/metabolism , Insulin Resistance , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Interleukin-6/metabolism , Interleukin-6/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , PPAR gamma/metabolism , Phosphorylation , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type II/agonists , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , NF-kappaB-Inducing Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...