Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
Braz. J. Pharm. Sci. (Online) ; 59: e21494, 2023. tab, graf
Article in English | LILACS | ID: biblio-1439544

ABSTRACT

Abstract Acute pancreatitis (AP) is a life-unpleasant situation with contradictory and inadequate treatments. In this regard, the present study evaluated the effect of the possible pretreatment of lipase-pancreatin on L-arginine-induced AP. Forty adult mice were selected and divided into five groups: I) control group, II and III) AP groups (i.p.) receiving L-arginine of 2×300 and 2×400 mg/100 g body weight (b.w.), IV) AP (2×300 L-arginine) group + pancreatin (mice were i.p. injected by 350 U-lipase), and V) AP (2×400 L-arginine) group + pancreatin (mice were i.p. injected by 350 U-lipase). All AP groups displayed a significant increase in serum levels of ALT, AST, TBARS, and TNF-alpha compared to the control group. Moreover, pancreatic tissue edema, inflammation, and vacuolization of acinar cells were significantly higher in the untreated L-arginine group compared to the control and pancreatin groups. Conversely, the diameter of pancreatic islets significantly declined after induction of pancreatitis compared with control and pancreatin groups. Pancreatin treatment can be used in pancreatic dysfunction, however, this medicine showed no protective effect against L-arginine-induced AP in the mouse model.


Subject(s)
Animals , Male , Mice , Pancreatitis/chemically induced , Pancreatin/adverse effects , Tumor Necrosis Factor-alpha/agonists , Acinar Cells/classification
3.
Am J Physiol Renal Physiol ; 320(6): F1159-F1164, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33969695

ABSTRACT

Although administration of hypertonic saline (HSS) in combination with diuretics has yielded improved weight loss, preservation of renal function, and reduction in hospitalization time in the clinical setting of patients with acute decompensated heart failure (ADHF), the mechanisms that underlie these beneficial effects remain unclear and additional studies are needed before this approach can be adopted on a more consistent basis. As high salt conditions stimulate the production of several renal autacoids that exhibit natriuretic effects, renal physiologists can contribute to the understanding of mechanisms by which HSS leads to increased diuresis both as an individual therapy as well as in combination with loop diuretics. For instance, since HSS increases TNF-α production by proximal tubule and thick ascending limb of Henle's loop epithelial cells, this article is aimed at highlighting how the effects of TNF-α produced by these cell types may contribute to the beneficial effects of HSS in patients with ADHF. Although TNF-α produced by infiltrating macrophages and T cells exacerbates and attenuates renal damage, respectively, production of this cytokine within the tubular compartment of the kidney functions as an intrinsic regulator of blood pressure and Na+ homeostasis via mechanisms along the nephron related to inhibition of Na+-K+-2Cl- cotransporter isoform 2 activity and angiotensinogen expression. Thus, in the clinical setting of ADHF and hyponatremia, induction of TNF-α production along the nephron by administration of HSS may attenuate Na+-K+-2Cl- cotransporter isoform 2 activity and angiotensinogen expression as part of a mechanism that prevents excessive Na+ reabsorption in the thick ascending limb of Henle's loop, thereby mitigating volume overload.


Subject(s)
Heart Failure/drug therapy , Saline Solution, Hypertonic/pharmacology , Tumor Necrosis Factor-alpha/agonists , Diuretics/therapeutic use , Gene Expression Regulation/drug effects , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Solute Carrier Family 12, Member 1/genetics , Solute Carrier Family 12, Member 1/metabolism
4.
J Biol Chem ; 296: 100220, 2021.
Article in English | MEDLINE | ID: mdl-33839684

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is an important transcription factor involved in many physiological functions including embryonic development and immune responses and is often activated under pathological conditions such as cancer. Strategies to inactivate STAT3 are being pursued as potential anticancer therapies and have led to the identification of Stattic (6-nitrobenzo[b]thiophene-1,1-dioxide) as a "specific" STAT3 inhibitor that is often used to interrogate STAT3-mediated gene expression in vitro and in vivo. Here, we show that Stattic exerts many STAT3-independent effects on cancer cells, calling for reassessment of results previously ascribed to STAT3 functions. Studies of the STAT3-deficient prostate cancer cell line PC-3 (PC3) along with STAT3-proficient breast cancer cell lines (MDA-MB-231, SUM149) revealed that Stattic attenuated histone acetylation and neutralized effects of the histone deacetylase (HDAC) inhibitor romidepsin. In PC3 cells, Stattic alone inhibited gene expression of CCL20 and CCL2, but activated expression of TNFA, CEBPD, SOX2, and MYC. In addition, we found that Stattic promoted autophagy and caused cell death. These data point to profound epigenetic effects of Stattic that are independent of its function as a STAT3 inhibitor. Our results demonstrate that Stattic directly or indirectly reduces histone acetylation and suggest reevaluation of Stattic and related compounds as polypharmacological agents through multipronged cytotoxic effects on cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclic S-Oxides/pharmacology , Gene Expression Regulation, Neoplastic , Histones/genetics , Protein Processing, Post-Translational , STAT3 Transcription Factor/genetics , Acetylation/drug effects , Autophagy/drug effects , Autophagy/genetics , CCAAT-Enhancer-Binding Protein-delta/agonists , CCAAT-Enhancer-Binding Protein-delta/genetics , CCAAT-Enhancer-Binding Protein-delta/metabolism , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL20/antagonists & inhibitors , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histones/antagonists & inhibitors , Histones/metabolism , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , PC-3 Cells , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-myc/agonists , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/agonists , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Red Fluorescent Protein
5.
Int J Biol Macromol ; 171: 177-184, 2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33421465

ABSTRACT

A water-soluble polysaccharide (LCP-05) was isolated from the flowers of Leucosceptrum canum Smith. LCP-05 was an acidic polysaccharide with a molecular weight of approximately 8.9 kDa. Monosaccharide composition analysis indicated that LCP-05 was composed of Man, Rha, GlcA, GalA, Glc, Gal and Ara in a molar ratio of 0.83:1.68:0.33:2.15:1.00:1.45:1.22. The framework of LCP-05 was speculated to be a branched rhamnogalacturonan with the backbone consisting of α-1,2,4-linked Rhap and α-1,4-linked GalAp, and bearing branches at the O-4 position of the Rha residues. The side chains are terminated primarily with the Araf and Glcp residues. LCP-05 was found to be able to significantly induce the production of NO, IL-6, and TNF-α in RAW 264.7 cells, and to induce RAW 264.7 cell's suppressive effect on both cell growth and cell migration of 4 T1 mammary breast cancer cells.


Subject(s)
Epithelial Cells/drug effects , Immunologic Factors/pharmacology , Lamiaceae/chemistry , Polysaccharides/pharmacology , Animals , Carbohydrate Sequence , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial Cells/pathology , Flowers/chemistry , Humans , Immunologic Factors/chemistry , Immunologic Factors/isolation & purification , Interleukin-6/agonists , Interleukin-6/immunology , Mice , Molecular Weight , Monosaccharides/chemistry , Monosaccharides/isolation & purification , Nitric Oxide/agonists , Nitric Oxide/immunology , Plant Extracts/chemistry , Polysaccharides/chemistry , Polysaccharides/isolation & purification , RAW 264.7 Cells , Solubility , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/immunology
6.
Int J Mol Sci ; 21(22)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33228049

ABSTRACT

The inhalation of metal (including lead) nanoparticles poses a real health issue to people and animals living in polluted and/or industrial areas. In this study, we exposed mice to lead(II) nitrate nanoparticles [Pb(NO3)2 NPs], which represent a highly soluble form of lead, by inhalation. We aimed to uncover the effects of their exposure on individual target organs and to reveal potential variability in the lead clearance. We examined (i) lead biodistribution in target organs using laser ablation and inductively coupled plasma mass spectrometry (LA-ICP-MS) and atomic absorption spectrometry (AAS), (ii) lead effect on histopathological changes and immune cells response in secondary target organs and (iii) the clearance ability of target organs. In the lungs and liver, Pb(NO3)2 NP inhalation induced serious structural changes and their damage was present even after a 5-week clearance period despite the lead having been almost completely eliminated from the tissues. The numbers of macrophages significantly decreased after 11-week Pb(NO3)2 NP inhalation; conversely, abundance of alpha-smooth muscle actin (α-SMA)-positive cells, which are responsible for augmented collagen production, increased in both tissues. Moreover, the expression of nuclear factor κB (NF-κB) and selected cytokines, such as tumor necrosis factor alpha (TNFα), transforming growth factor beta 1 (TGFß1), interleukin 6(IL-6), IL-1α and IL-1ß , displayed a tissue-specific response to lead exposure. In summary, diminished inflammatory response in tissues after Pb(NO3)2 NPs inhalation was associated with prolonged negative effect of lead on tissues, as demonstrated by sustained pathological changes in target organs, even after long clearance period.


Subject(s)
Air Pollutants/pharmacokinetics , Lead/pharmacokinetics , Lung/drug effects , Macrophages, Alveolar/drug effects , Metal Nanoparticles/toxicity , Nitrates/pharmacokinetics , Actins/agonists , Actins/genetics , Actins/immunology , Administration, Inhalation , Air Pollutants/toxicity , Animals , Biological Availability , Female , Gene Expression , Half-Life , Inhalation Exposure/analysis , Interleukin-1alpha/agonists , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-1beta/agonists , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/agonists , Interleukin-6/genetics , Interleukin-6/immunology , Lead/toxicity , Liver/drug effects , Liver/immunology , Liver/pathology , Lung/immunology , Lung/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Metal Nanoparticles/administration & dosage , Mice , Mice, Inbred ICR , NF-kappa B/agonists , NF-kappa B/genetics , NF-kappa B/immunology , Nitrates/toxicity , Spectrophotometry, Atomic , Tissue Distribution , Transforming Growth Factor beta1/agonists , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
7.
Bull Exp Biol Med ; 168(6): 767-772, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32328942

ABSTRACT

In 1 and 24 h after combined administration of TLR4 (LPS) and TLR3 (Poly I:C) ligands to CBA mice, the content of MSC in bone marrow increased to intermediate value between the levels attained by their individual injections. The content of osteogenic MSC assessed in 24 h postinjection corresponded to the control level in Poly I:C group, decreased in LPS group by 2.5 times relatively to the control, and increased by 1.6 times (relatively to control) after combined administration of the ligands. In 3 h after combined addition of LPS and Poly I:C in vitro to 12-day-old primary culture of mouse bone marrow stromal cells, the concentration of TNFα in culture medium was intermediate between the levels attained by their individual application. The data revealed dependence of activation of stromal tissue on intensity of innate immunity reactions; they also attested to marked elevation of osteogenicity of MSC pool after costimulation with Poly I:C and LPS, which can underlie augmented calcification of the tissues during combined viral and bacterial infections.


Subject(s)
Lipopolysaccharides/pharmacology , Mesenchymal Stem Cells/drug effects , Poly I-C/pharmacology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 4/genetics , Animals , Cell Count , Cell Differentiation/drug effects , Drug Synergism , Gene Expression , Immunity, Innate/drug effects , Injections, Intraperitoneal , Interleukin-10/agonists , Interleukin-10/genetics , Interleukin-10/immunology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred CBA , Osteogenesis/drug effects , Osteogenesis/genetics , Osteogenesis/immunology , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
8.
J Med Econ ; 22(9): 859-868, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31012362

ABSTRACT

Aims: To evaluate the cost differences between a treatment strategy including tofacitinib (TOFA) vs treatment strategies including adalimumab (ADA), golimumab (GOL), infliximab (IFX), and vedolizumab (VEDO) among all patients with moderate-to-severe ulcerative colitis (UC) (further stratified by patients naïve/exposed to tumor necrosis factor inhibitors [TNFis]). Materials and methods: An Excel-based decision-analytic model was developed to evaluate costs from the perspective of a third-party US payer over 2 years. Efficacy and safety parameters were taken from prescribing information and published trials. All patients started induction therapy on the first treatment in the strategy and continued if efficacy criteria were met and no major adverse event occurred (in which cases they proceeded to the next treatment in the strategy). Results: The cost per member per month (PMPM) of the TOFA->IFX->VEDO->GOL strategy ($1.11) was lower than that of the ADA->IFX->VEDO->GOL strategy ($1.34; Δ = $-0.23) among the TNFi-naïve population (n = 204 patients out of a plan of one million members). Similarly, the use of TOFA before ADA (i.e. TOFA->ADA->IFX-> VEDO) was also associated with lower PMPM costs than the use of ADA before TOFA (i.e. ADA->TOFA->IFX->VEDO): $1.15 vs $1.25 (Δ = $-0.10). Similar, and often larger, differences were observed in both the overall moderate-to-severe population and the TNFi-exposed population. Sensitivity analyses resulted in the same conclusions. Limitations: Our model relied on efficacy data from prescribing information and published trials, which were not head-to-head and slightly differed with respect to methods. Additionally, our model used representative minor and major ADRs (and the associated costs) to represent toxicity management, which was a simplifying assumption. Conclusions: This analysis, the first of its kind to evaluate TOFA vis-à-vis other advanced therapies in the US, suggests the early use of TOFA among both TNFi-naïve and TNFi-failure patients results in lower PMPM costs compared with other treatment alternatives.


Subject(s)
Colitis, Ulcerative/drug therapy , Gastrointestinal Agents/economics , Gastrointestinal Agents/therapeutic use , Health Expenditures/statistics & numerical data , Piperidines/economics , Piperidines/therapeutic use , Pyrimidines/economics , Pyrimidines/therapeutic use , Pyrroles/economics , Pyrroles/therapeutic use , Adalimumab/economics , Adalimumab/therapeutic use , Antibodies, Monoclonal/economics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/economics , Antibodies, Monoclonal, Humanized/therapeutic use , Cost-Benefit Analysis , Gastrointestinal Agents/adverse effects , Health Resources/economics , Health Resources/statistics & numerical data , Health Services/economics , Health Services/statistics & numerical data , Humans , Infliximab/economics , Infliximab/therapeutic use , Models, Econometric , Patient Acceptance of Health Care/statistics & numerical data , Piperidines/adverse effects , Pyrimidines/adverse effects , Pyrroles/adverse effects , Severity of Illness Index , Tumor Necrosis Factor-alpha/agonists , United States
9.
J Physiol Biochem ; 74(4): 579-589, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29790117

ABSTRACT

The main aim of this investigation was to study the regulatory roles of let-7b and miR-155-3p on the expression of inflammation-associated genes in monocytes, macrophages, and lipopolysaccharide (LPS)-activated macrophages (AcM). A second goal was to analyze the potential modulatory roles of different fatty acids, including oleic, palmitic, eicosapentaenoic (EPA), and docosahexaenoic (DHA), on the expression of these miRNAs in the three cell types. This hypothesis was tested in human acute monocytic leukemia cells (THP-1), which were differentiated into macrophages with 2-O-tetradecanoylphorbol-13-acetate (TPA) and further activated with LPS for 24 h. Monocytes, macrophages, and AcM were transfected with a negative control, or mimics for miR-155-3p and miR-let-7b-5p. The expression of both miRNAs and some proinflammatory genes was analyzed by qRT-PCR. Interestingly, let-7b mimic reduced the expression of IL6 and TNF in monocytes, and SERPINE1 expression in LPS-activated macrophages. However, IL6, TNF, and SERPINE1 were upregulated in macrophages by let-7b mimic. IL6 expression was higher in the three types of cells after transfecting with miR-155-3p mimic. Similarly, expression of SERPINE1 was increased by miR-155-3p mimic in monocytes and macrophages. However, TLR4 was downregulated by miR-155-3p in monocytes and macrophages. Regarding the effects of the different fatty acids, oleic acid increased the expression of let-7b in macrophages and AcM and also increased the expression of miR-155 in monocytes when compared with DHA but not when compared with non-treated cells. Overall, these results suggest anti- and proinflammatory roles of let-7b and miR-155-3p in THP-1 cells, respectively, although these outcomes are strongly dependent on the cell type. Noteworthy, oleic acid might exert beneficial anti-inflammatory effects in immune cells (i.e., non-activated and LPS-activated macrophages) by upregulating the expression of let-7b.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Gene Expression Regulation , Macrophages/metabolism , MicroRNAs/metabolism , Monocytes/metabolism , Cell Differentiation/drug effects , Docosahexaenoic Acids/metabolism , Down-Regulation , Gene Expression Regulation/drug effects , Humans , Interleukin-6/agonists , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , MicroRNAs/chemistry , Monocytes/immunology , Oleic Acid/metabolism , Plasminogen Activator Inhibitor 1/agonists , Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , RNA/metabolism , THP-1 Cells , Tetradecanoylphorbol Acetate/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
Arch Physiol Biochem ; 124(5): 468-474, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29364751

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the effects of high glucose (HG) on inflammation in HepG2 cells. METHODS: The molecular mechanisms linking HG to inflammation was assessed in HepG2 cells exposed to HG (33 mM). RESULTS: The results showed that HG significantly enhanced TNF-α, IL-6 and PAI-1 expression in HepG2 cells. Increased expression of cytokines was accompanied by enhanced phosphorylation of JNK, P38, ERK and IKKα/IKKß. In addition, JNK, ERK, P38 and NF-kB inhibitors could significantly attenuate HG-induced expression of TNF-α, IL-6 and PAI-1. Furthermore, HG could promote the generation of reactive oxygen species (ROS), while N-acetyl cysteine, a ROS scavenger, had an inhibitory effect on the expression of TNF-α, IL-6 and PAI-1 in HG-treated cells. CONCLUSIONS: Our results indicated that HG-induced inflammation is mediated through the generation of ROS and activation of the MAPKs and NF-kB signalling pathways in HepG2 cells.


Subject(s)
Gene Expression Regulation , Hepatocytes/metabolism , Hyperglycemia/metabolism , MAP Kinase Signaling System , NF-kappa B/agonists , Oxidative Stress , Tumor Necrosis Factor-alpha/agonists , Acetylcysteine/pharmacology , Free Radical Scavengers/pharmacology , Gene Expression Regulation/drug effects , Glucose/adverse effects , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/immunology , Hepatocytes/pathology , Humans , Hyperglycemia/immunology , Hyperglycemia/pathology , Interleukin-6/agonists , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/metabolism , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Osmolar Concentration , Oxidative Stress/drug effects , Phosphorylation/drug effects , Plasminogen Activator Inhibitor 1/agonists , Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Reproducibility of Results , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
11.
J Biol Chem ; 292(33): 13714-13726, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28646109

ABSTRACT

Bone morphogenetic proteins 9 and 10 (BMP9/BMP10) are circulating cytokines with important roles in endothelial homeostasis. The aim of this study was to investigate the roles of BMP9 and BMP10 in mediating monocyte-endothelial interactions using an in vitro flow adhesion assay. Herein, we report that whereas BMP9/BMP10 alone had no effect on monocyte recruitment, at higher concentrations both cytokines synergized with tumor necrosis factor-α (TNFα) to increase recruitment to the vascular endothelium. The BMP9/BMP10-mediated increase in monocyte recruitment in the presence of TNFα was associated with up-regulated expression levels of E-selectin, vascular cell adhesion molecule (VCAM-1), and intercellular adhesion molecule 1 (ICAM-1) on endothelial cells. Using siRNAs to type I and II BMP receptors and the signaling intermediaries (Smads), we demonstrated a key role for ALK2 in the BMP9/BMP10-induced surface expression of E-selectin, and both ALK1 and ALK2 in the up-regulation of VCAM-1 and ICAM-1. The type II receptors, BMPR-II and ACTR-IIA were both required for this response, as was Smad1/5. The up-regulation of cell surface adhesion molecules by BMP9/10 in the presence of TNFα was inhibited by LDN193189, which inhibits ALK2 but not ALK1. Furthermore, LDN193189 inhibited monocyte recruitment induced by TNFα and BMP9/10. BMP9/10 increased basal IκBα protein expression, but did not alter p65/RelA levels. Our findings suggest that higher concentrations of BMP9/BMP10 synergize with TNFα to induce the up-regulation of endothelial selectins and adhesion molecules, ultimately resulting in increased monocyte recruitment to the vascular endothelium. This process is mediated mainly via the ALK2 type I receptor, BMPR-II/ACTR-IIA type II receptors, and downstream Smad1/5 signaling.


Subject(s)
Activin Receptors, Type I/metabolism , Bone Morphogenetic Proteins/metabolism , Endothelium, Vascular/metabolism , Growth Differentiation Factors/metabolism , Monocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Activin Receptors, Type I/antagonists & inhibitors , Activin Receptors, Type I/genetics , Activin Receptors, Type II/antagonists & inhibitors , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Aorta , Cell Adhesion/drug effects , Cells, Cultured , E-Selectin/chemistry , E-Selectin/genetics , E-Selectin/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Growth Differentiation Factor 2 , Humans , Intercellular Adhesion Molecule-1/chemistry , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Kinetics , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA Interference , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/agonists , Up-Regulation/drug effects , Vascular Cell Adhesion Molecule-1/chemistry , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
12.
J Biol Chem ; 292(24): 10169-10179, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28438834

ABSTRACT

Cytokines, including receptor activator of nuclear factor κB ligand (RANKL) and TNF, induce increased osteoclast (OC) formation and bone loss in postmenopausal osteoporosis and inflammatory arthritides. RANKL and TNF can independently induce OC formation in vitro from WT OC precursors via TNF receptor-associated factor (TRAF) adaptor proteins, which bind to their receptors. Of these, only TRAF6 is required for RANKL-induced osteoclastogenesis in vitro However, the molecular mechanisms involved remain incompletely understood. Here we report that RANKL induced the formation of bone-resorbing OCs from TRAF6-/- OC precursors when cultured on bone slices but not on plastic. The mechanisms involved increased TNF production by TRAF6-/- OC precursors resulting from their interaction with bone matrix and release of active TGFß from the resorbed bone, coupled with RANKL-induced autophagolysosomal degradation of TRAF3, a known inhibitor of OC formation. Consistent with these findings, RANKL enhanced TNF-induced OC formation from TRAF6-/- OC precursors. Moreover, TNF induced significantly more OCs from mice with TRAF3 conditionally deleted in myeloid lineage cells, and it did not inhibit RANKL-induced OC formation from these cells. TRAF6-/- OC precursors that overexpressed TRAF3 or were treated with the autophagolysosome inhibitor chloroquine formed significantly fewer OCs in response to TNF alone or in combination with RANKL. We conclude that RANKL can enhance TNF-induced OC formation independently of TRAF6 by degrading TRAF3. These findings suggest that preventing TRAF3 degradation with drugs like chloroquine could reduce excessive OC formation in diseases in which bone resorption is increased in response to elevated production of these cytokines.


Subject(s)
Bone and Bones/metabolism , Osteoclasts/metabolism , Osteogenesis , RANK Ligand/metabolism , TNF Receptor-Associated Factor 3/metabolism , TNF Receptor-Associated Factor 6/metabolism , Tumor Necrosis Factor-alpha/agonists , Actin Cytoskeleton/immunology , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Autophagy/drug effects , Bone Demineralization, Pathologic/immunology , Bone Demineralization, Pathologic/metabolism , Bone Demineralization, Pathologic/pathology , Bone and Bones/drug effects , Bone and Bones/immunology , Bone and Bones/pathology , Cell Culture Techniques , Cells, Cultured , Chloroquine/pharmacology , Mice , Mice, Knockout , Osteoclasts/drug effects , Osteoclasts/immunology , Osteoclasts/pathology , Osteogenesis/drug effects , Protein Stability/drug effects , Proteolysis/drug effects , RANK Ligand/genetics , Spleen/cytology , Spleen/immunology , Spleen/metabolism , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 6/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
13.
Article in English | MEDLINE | ID: mdl-28167544

ABSTRACT

Visceral leishmaniasis is a fatal parasitic neglected disease affecting 1.5 million people worldwide. Based on a drug repositioning approach, the aim of this work was to investigate the in vitro immunomodulatory potential of buparvaquone (BPQ) and to establish a safe regimen to evaluate the in vivo efficacy of BPQ entrapped by negatively charged nanoliposomes (BPQ-LP) in Leishmania infantum-infected hamsters. Small-angle X-ray scattering, dynamic light scattering, and the ζ-potential were applied in order to study the influence of BPQ on the liposome structure. Our data revealed that BPQ was located in the polar-apolar interface, snorkeling the polar region, and protected against aggregation inside the lipophilic region. The presence of BPQ also decreased the Z-average hydrodynamic diameter and increased the surface charge. Compared to intravenous and intramuscular administration, a subcutaneous route was a more effective route for BPQ-LP; at 0.4 mg/kg, BPQ-LP reduced infection in the spleen and liver by 98 and 96%, respectively. Treatment for 5 days resulted in limited efficacy, but 10 days of treatment resulted in an efficacy similar to that of a 15-day regimen. The nanoliposomal drug was highly effective, with a mean 50% effective dose of 0.25 mg/kg, reducing the parasite load in bone marrow by 80%, as detected using quantitative PCR analysis. In addition, flow cytometry studies showed that BPQ upregulated cytokines as tumor necrosis factor, monocyte chemoattractant protein 1, interleukin-10 (IL-10), and IL-6 in Leishmania-infected macrophages, eliminating the parasites via a nitric oxide-independent mechanism. This new formulation proved to be a safe and effective treatment for murine leishmaniasis that could be a useful candidate against visceral leishmaniasis.


Subject(s)
Antiprotozoal Agents/pharmacology , Immunologic Factors/pharmacology , Leishmania infantum/drug effects , Leishmaniasis, Visceral/drug therapy , Liposomes/chemistry , Macrophages/drug effects , Naphthoquinones/pharmacology , Administration, Cutaneous , Animals , Antiprotozoal Agents/chemistry , Bone Marrow/drug effects , Bone Marrow/immunology , Bone Marrow/parasitology , Chemokine CCL2/agonists , Chemokine CCL2/biosynthesis , Cricetinae , Disease Models, Animal , Drug Compounding/methods , Immunologic Factors/chemistry , Interleukin-10/agonists , Interleukin-10/biosynthesis , Interleukin-6/agonists , Interleukin-6/biosynthesis , Leishmania infantum/growth & development , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Liposomes/pharmacokinetics , Liver/drug effects , Liver/immunology , Liver/parasitology , Macrophages/immunology , Macrophages/parasitology , Male , Mice , Nanostructures/administration & dosage , Nanostructures/chemistry , Naphthoquinones/chemistry , Parasite Load , Spleen/drug effects , Spleen/immunology , Spleen/parasitology , Static Electricity , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/biosynthesis
14.
J Physiol Biochem ; 73(2): 287-296, 2017 May.
Article in English | MEDLINE | ID: mdl-28181168

ABSTRACT

Endothelial dysfunction plays a vital role during the initial stage of atherosclerosis. Oxidized low-density lipoprotein (ox-LDL) induces vascular endothelial injury and vessel wall inflammation. Sphingosine-1-phosphate (S1P) exerts numerous vasoprotective effects by binding to diverse S1P receptors (S1PRs; S1PR1-5). A number of studies have shown that in endothelial cells (ECs), S1PR2 acts as a pro-atherosclerotic mediator by stimulating vessel wall inflammation through the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. Scavenger receptor class B member I (SR-BI), a high-affinity receptor for apolipoprotein A-I (apoA-I)/high-density lipoprotein (HDL), inhibits nuclear factor-κB (NF-κB) translocation and decreases the plasma levels of inflammatory mediators via the PI3K/Akt pathway. We hypothesized that the inflammatory effects of S1P/S1PR2 on ECs may be regulated by apoA-I/SR-BI. The results showed that ox-LDL, a pro-inflammatory factor, augmented the S1PR2 level in human umbilical vein endothelial cells (HUVECs) in a dose- and time-dependent manner. In addition, S1P/S1PR2 signaling influenced the levels of inflammatory factors, including tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and IL-10, aggravating inflammation in HUVECs. Moreover, the pro-inflammatory effects induced by S1P/S1PR2 were attenuated by SR-BI overexpression and enhanced by an SR-BI inhibitor, BLT-1. Further experiments showed that the PI3K/Akt signaling pathway was involved in this process. Taken together, these results demonstrate that apoA-I/SR-BI negatively regulates S1P/S1PR2-mediated inflammation in HUVECs by activating the PI3K/Akt signaling pathway.


Subject(s)
Apolipoprotein A-I/metabolism , Endothelium, Vascular/metabolism , Lysophospholipids/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Receptors, Lysosphingolipid/agonists , Scavenger Receptors, Class B/agonists , Signal Transduction , Sphingosine/analogs & derivatives , Active Transport, Cell Nucleus/drug effects , Apolipoprotein A-I/genetics , Cells, Cultured , Cyclopentanes/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-10/agonists , Interleukin-10/metabolism , Interleukin-1beta/agonists , Interleukin-1beta/metabolism , Kinetics , Lipoproteins, LDL/adverse effects , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Scavenger Receptors, Class B/antagonists & inhibitors , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Signal Transduction/drug effects , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors , Thiosemicarbazones/pharmacology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/metabolism
15.
Arch Toxicol ; 91(1): 231-246, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27052459

ABSTRACT

Astrocytes, the largest cell population in the human brain, are powerful inflammatory effectors. Several studies have examined the interaction of activated astrocytes with neurons, but little is known yet about human neurotoxicity under such situations and about strategies of neuronal rescue. To address this question, immortalized murine astrocytes (IMA) were combined with human LUHMES neurons and stimulated with an inflammatory (TNF, IL-1) cytokine mix (CM). Neurotoxicity was studied both in co-cultures and in monocultures after transfer of conditioned medium from activated IMA. Interventions with >20 drugs were used to profile the model system. Control IMA supported neurons and protected them from neurotoxicants. Inflammatory activation reduced this protection, and prolonged exposure of co-cultures to CM triggered neurotoxicity. Neither the added cytokines nor the release of NO from astrocytes were involved in this neurodegeneration. The neurotoxicity-mediating effect of IMA was faithfully reproduced by human astrocytes. Moreover, glia-dependent toxicity was also observed, when IMA cultures were stimulated with CM, and the culture medium was transferred to neurons. Such neurotoxicity was prevented when astrocytes were treated by p38 kinase inhibitors or dexamethasone, whereas such compounds had no effect when added to neurons. Conversely, treatment of neurons with five different drugs, including resveratrol and CEP1347, prevented toxicity of astrocyte supernatants. Thus, the sequential IMA-LUHMES neuroinflammation model is suitable for separate profiling of both glial-directed and directly neuroprotective strategies. Moreover, direct evaluation in co-cultures of the same cells allows for testing of therapeutic effectiveness in more complex settings, in which astrocytes affect pharmacological properties of neurons.


Subject(s)
Astrocytes/drug effects , Cell Communication/drug effects , Cytokines/agonists , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Astrocytes/cytology , Astrocytes/immunology , Astrocytes/metabolism , Cell Line, Transformed , Cell Survival/drug effects , Coculture Techniques , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Dexamethasone/antagonists & inhibitors , Dexamethasone/pharmacology , Hormone Antagonists/pharmacology , Humans , Interleukin-1beta/agonists , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mifepristone/pharmacology , Neurogenesis/drug effects , Neuronal Plasticity/drug effects , Neurons/cytology , Neurons/immunology , Neurons/metabolism , Neurotoxins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
16.
Sci Rep ; 6: 38294, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27910896

ABSTRACT

Methylmercury selectively damages the central nervous system (CNS). The tumor necrosis factor (TNF) superfamily includes representative cytokines that participate in the inflammatory response as well as cell survival, and apoptosis. In this study, we found that administration of methylmercury selectively induced TNF-α expression in the brain of mice. Although the accumulated mercury concentration in the liver and kidneys was greater than in the brain, TNF-α expression was induced to a greater extent in brain. Thus, it is possible that there may exist a selective mechanism by which methylmercury induces TNF-α expression in the brain. We also found that TNF-α expression was induced by methylmercury in C17.2 cells (mouse neural stem cells) and NF-κB may participate as a transcription factor in that induction. Further, we showed that the addition of TNF-α antagonist (WP9QY) reduced the toxicity of methylmercury to C17.2 cells. In contrast, the addition of recombinant TNF-α to the culture medium decreased the cell viability. We suggest that TNF-α may play a part in the selective damage of the CNS by methylmercury. Furthermore, our results indicate that the higher TNF-α expression induced by methylmercury maybe the cause of cell death, as TNF-α binds to its receptor after being released extracellularly.


Subject(s)
Brain/drug effects , Methylmercury Compounds/toxicity , Neural Stem Cells/drug effects , Tumor Necrosis Factor-alpha/genetics , Animals , Antidotes/pharmacology , Brain/metabolism , Brain/pathology , Cell Death/drug effects , Cell Line , Gene Expression Profiling , Gene Expression Regulation , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Organ Specificity , Peptides, Cyclic/pharmacology , Protein Binding , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
17.
Sci Rep ; 6: 27566, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27297123

ABSTRACT

UNLABELLED: Chemical regulation of macrophage function is one key strategy for developing host-directed adjuvant therapies for tuberculosis (TB). A critical step to develop these therapies is the identification and characterization of specific macrophage molecules and pathways with a high potential to serve as drug targets. Using a barcoded lentivirus-based pooled short-hairpin RNA (shRNA) library combined with next generation sequencing, we identified 205 silenced host genes highly enriched in mycobacteria-resistant macrophages. Twenty-one of these "hits" belonged to the oxidoreductase functional category. NAD(P)H: quinone oxidoreductase 1 (NQO1) was the top oxidoreductase "hit". NQO1 expression was increased after mycobacterial infection, and NQO1 knockdown increased macrophage differentiation, NF-κB activation, and the secretion of pro-inflammatory cytokines TNF-α and IL-1ß in response to infection. This suggests that mycobacteria hijacks NQO1 to down-regulate pro-inflammatory and anti-bacterial functions. The competitive inhibitor of NQO1 dicoumarol synergized with rifampin to promote intracellular killing of mycobacteria. Thus, NQO1 is a new host target in mycobacterial infection that could potentially be exploited to increase antibiotic efficacy in vivo. Our findings also suggest that pooled shRNA libraries could be valuable tools for genome-wide screening in the search for novel druggable host targets for adjunctive TB therapies.


Subject(s)
Antitubercular Agents/pharmacology , Dicumarol/pharmacology , Host-Pathogen Interactions/drug effects , Macrophages/immunology , Mycobacterium tuberculosis/drug effects , NAD(P)H Dehydrogenase (Quinone)/genetics , Drug Synergism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Gene Library , High-Throughput Nucleotide Sequencing , High-Throughput Screening Assays , Humans , Interleukin-1beta/agonists , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Macrophages/drug effects , Macrophages/microbiology , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/physiology , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/immunology , NF-kappa B/agonists , NF-kappa B/genetics , NF-kappa B/immunology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rifampin/pharmacology , Signal Transduction , THP-1 Cells , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
18.
Toxicol Lett ; 258: 36-45, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27313094

ABSTRACT

Inflammatory responses are involved in mechanisms of neuronal cell damage in the pathogenesis of neurodegenerative diseases such as Parkinson's disease (PD). We investigated the mechanisms whereby inflammatory responses contribute to loss of dopaminergic neurons in fipronil (FPN)-treated rats. After stereotaxic injection of FPN in the substantia nigra (SN), the number of tyrosine hydroxylase (TH)-positive neurons and the levels of TH expression in the SN decreased at 7days, and a significant decrease was observed at 14days with a subsequent reduction in striatal TH expression. Decreases in dopamine (DA) levels, however, began at 3days post-injection, preceding the changes in TH expression. In contrast, glial fibrillary acidic protein (GFAP) expression was significantly increased at 3days and persisted for up to 14days post-lesion; these changes in GFAP expression appeared to be inversely correlated with TH expression. Furthermore, we found that FPN administration induced an inflammatory response characterized by increased levels of inducible NO synthase (iNOS), cyclooxygenase-2 (COX-2), and tumor necrosis factor-α (TNF-α), which was mediated by activated microglia following infusion of FPN unilaterally into the SN. Intranigral injection of FPN underwent an inflammatory response with a resultant ongoing loss of dopaminergic neurons, indicating that pesticides may have important implication for the study of PD.


Subject(s)
Dopaminergic Neurons/drug effects , Encephalitis/etiology , Neurotoxicity Syndromes/pathology , Pesticides/toxicity , Pyrazoles/toxicity , Striatonigral Degeneration/etiology , Substantia Nigra/drug effects , Animals , Biomarkers/metabolism , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/metabolism , Disease Progression , Dopamine/chemistry , Dopamine/metabolism , Dopaminergic Neurons/immunology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Glial Fibrillary Acidic Protein/agonists , Glial Fibrillary Acidic Protein/metabolism , Injections, Intraventricular , Male , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/physiopathology , Nitric Oxide Synthase Type II/chemistry , Nitric Oxide Synthase Type II/metabolism , Pyrazoles/administration & dosage , Random Allocation , Rats, Sprague-Dawley , Stereotaxic Techniques , Substantia Nigra/immunology , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/metabolism , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Tyrosine 3-Monooxygenase/metabolism
19.
Biosci Biotechnol Biochem ; 80(7): 1393-402, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27095137

ABSTRACT

We herein report the immunostimulatory effect of spinach aqueous extract (SAE) on mouse macrophage-like J774.1 cells and mouse primary peritoneal macrophages. SAE significantly enhanced the production of interleukin (IL)-6 and tumor necrosis factor-α by both J774.1 cells and peritoneal macrophages by enhancing the expression levels of these cytokine genes. In addition, the phagocytosis activity of J774.1 cells was facilitated by SAE. Immunoblot analysis revealed that SAE activates mitogen-activated protein kinase and nuclear factor-κB cascades. It was found that SAE activates macrophages through not only TLR4, but also other receptors. The production of IL-6 was significantly enhanced by peritoneal macrophages from SAE-administered BALB/c mice, suggesting that SAE has a potential to stimulate macrophage activity in vivo. Taken together, these data indicate that SAE would be a beneficial functional food with immunostimulatory effects on macrophages.


Subject(s)
Adjuvants, Immunologic/pharmacology , Interleukin-6/agonists , Macrophages, Peritoneal/drug effects , Plant Extracts/pharmacology , Spinacia oleracea/chemistry , Tumor Necrosis Factor-alpha/agonists , Animals , Cell Line , Female , Gene Expression , Interleukin-6/genetics , Interleukin-6/immunology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/immunology , NF-kappa B/agonists , NF-kappa B/genetics , NF-kappa B/immunology , Phagocytosis/drug effects , Primary Cell Culture , RNA, Messenger/agonists , RNA, Messenger/genetics , RNA, Messenger/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
20.
Biochem Pharmacol ; 116: 1-10, 2016 09 15.
Article in English | MEDLINE | ID: mdl-26993379

ABSTRACT

Tumor Necrosis Factor (TNF) is a potent inflammatory cytokine that exerts its functions through the activation of two distinct receptors, TNFR1 and TNFR2. Both receptors can activate canonical NF-κB and JNK MAP kinase signaling, while TNFR2 can also activate non-canonical NF-κB signaling, leading to numerous changes in gene expression that drive inflammation, cell proliferation and cell survival. On the other hand, TNFR1 also activates signaling pathways leading to cell death by either apoptosis or necroptosis, depending on the cellular context. A key player in TNFR1- and TNFR2-induced signaling is the RING finger protein TRAF2, which is recruited to both receptors upon their stimulation. TRAF2 exerts multiple receptor-specific functions but also mediates cross-talk between TNFR1 and TNFR2, dictating the outcome of TNF stimulation. In this review, we provide an overview of the positive and negative regulatory role of TRAF2 in different TNFR1 and TNFR2 signaling pathways. We discuss the underlying molecular mechanism of action, distinguishing between TRAF2 scaffold and E3 ubiquitin ligase functions, and the regulation of TRAF2 by specific post-translational modifications. Finally, we elaborate on some possible strategies to modulate TRAF2 function in the context of therapeutic targeting in autoimmunity and cancer.


Subject(s)
Apoptosis , Models, Biological , Necrosis/metabolism , Receptors, Tumor Necrosis Factor, Type II/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Signal Transduction , TNF Receptor-Associated Factor 2/metabolism , Animals , Humans , MAP Kinase Signaling System , NF-kappa B/metabolism , Phosphorylation , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Protein Stability , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , TNF Receptor-Associated Factor 2/chemistry , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...