Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters

Publication year range
1.
Molecules ; 27(3)2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35164164

ABSTRACT

Interleukin-1 beta (IL-1ß) has diverse physiological functions and plays important roles in health and disease. In this report, we focus on its function in the production of pro-inflammatory cytokines, including IL-6 and IL-8, which are implicated in several autoimmune diseases and host defense against infection. IL-1ß activity is markedly dependent on the binding affinity toward IL-1 receptors (IL-1Rs). Several studies have been conducted to identify suitable small molecules that can modulate the interactions between 1L-1ß and 1L-1R1. Based on our previous report, where DPIE [2-(1,2-Diphenyl-1H-indol-3-yl)ethanamine] exhibited such modulatory activity, three types of DPIE derivatives were synthesized by introducing various substituents at the 1, 2, and 3 positions of the indole group in DPIE. To predict a possible binding pose in complex with IL-1R1, a docking simulation was performed. The effect of the chemicals was determined in human gingival fibroblasts (GFs) following IL-1ß induction. The DPIE derivatives affected different aspects of cytokine production. Further, a group of the derivatives enabled synergistic pro-inflammatory cytokine production, while another group caused diminished cytokine production compared to DPIE stimulation. Some groups displayed no significant difference after stimulation. These findings indicate that the modification of the indole site could modulate IL-1ß:IL1R1 binding affinity to reduce or enhance pro-inflammatory cytokine production.


Subject(s)
Cytokines/agonists , Cytokines/antagonists & inhibitors , Indoles/pharmacology , Inflammation Mediators/agonists , Inflammation Mediators/antagonists & inhibitors , Phenethylamines/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Cytokines/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Indoles/chemistry , Inflammation/drug therapy , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-1beta/agonists , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/metabolism , Phenethylamines/chemistry
2.
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
3.
J Biol Chem ; 292(29): 12077-12087, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28576828

ABSTRACT

The NLRP3 inflammasome is a multiprotein complex that regulates the activation of caspase-1 leading to the maturation of the proinflammatory cytokines IL-1ß and IL-18 and promoting pyroptosis. Classically, the NLRP3 inflammasome in murine macrophages is activated by the recognition of pathogen-associated molecular patterns and by many structurally unrelated factors. Understanding the precise mechanism of NLRP3 activation by such a wide array of stimuli remains elusive, but several signaling events, including cytosolic efflux and influx of select ions, have been suggested. Accordingly, several studies have indicated a role of anion channels in NLRP3 inflammasome assembly, but their direct involvement has not been shown. Here, we report that the chloride intracellular channel proteins CLIC1 and CLIC4 participate in the regulation of the NLRP3 inflammasome. Confocal microscopy and cell fractionation experiments revealed that upon LPS stimulation of macrophages, CLIC1 and CLIC4 translocated into the nucleus and cellular membrane. In LPS/ATP-stimulated bone marrow-derived macrophages (BMDMs), CLIC1 or CLIC4 siRNA transfection impaired transcription of IL-1ß, ASC speck formation, and secretion of mature IL-1ß. Collectively, our results demonstrate that CLIC1 and CLIC4 participate both in the priming signal for IL-1ß and in NLRP3 activation.


Subject(s)
Chloride Channels/metabolism , Inflammasomes/drug effects , Interleukin-1beta/agonists , Macrophage Activation/drug effects , Macrophages/drug effects , Mitochondrial Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line , Cells, Cultured , Chloride Channels/antagonists & inhibitors , Chloride Channels/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lipopolysaccharides/toxicity , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Protein Transport/drug effects , Pyroptosis/drug effects , RAW 264.7 Cells , RNA Interference , Signal Transduction/drug effects
4.
Georgian Med News ; (278): 50-55, 2018 May.
Article in Russian | MEDLINE | ID: mdl-29905545

ABSTRACT

The purpose of our study was a comparative analysis of the effect of dentures from various materials on the immunological and redox-dependent homeostasis of the oral cavity. We studied 60 patients with removable dentures made based on plastics Prothyl Hot, Vertex BasiQ 20 (differing by polymerization regime) and elastic thermoplastic polymer Perflex Flexi Nylon. The control group consisted of 15 volunteers with a practically healthy oral cavity, who did not use dentures. Saliva collected on an empty stomach in a glass tube without the use of a stimulator before the establishment of a denture and 3 days and 1 month after. The content of the protein P-53 in saliva determined by immunoenzymatic assay with use of "Cusabio" reagent. The cytokines (IL1ß, IL10) content in saliva was determined immunoenzymatic assay. To determine the redox balance in the saliva of patients, the lipoperoxydradicals content (LOO.) content (by EPR method, using the spin-labeled α-phenyl-tertbutylnitron (PBN) (SIGMA)) and the activity of antioxidant enzymes (catalase and SOD) (by spectrophotometry) studied. Statistical processing of the results was carried out using the software package SPSS (version 10.0). Results of analysis show that defects associated with a lack of teeth do not affect the immune and oxidative balance of the oral cavity, but contribute to the development of destructive changes in the oral cavity's soft tissues, which manifested by an increase in the content of the proapoptotic protein P-53 in the saliva. After establishment of a denture, the intensity of apoptosis in the oral cavity tissues reduced. Establishment of a denture induced development of an inflammatory reaction during the first days, the intensity of which gradually decreased and completely disappeared at the end of the first month of the observation (manifested by the normalization of the parameters of the immune balance and antioxidant system). Minimal traumatic effects observed during establishment of a denture made based on Perflex Flexi Nylon.


Subject(s)
Dental Materials/therapeutic use , Denture, Partial, Removable , Gene Expression Regulation/drug effects , Resins, Synthetic/therapeutic use , Saliva/drug effects , Case-Control Studies , Catalase/genetics , Catalase/metabolism , Denture Design , Humans , Interleukin-10/agonists , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-1beta/agonists , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lipid Peroxides/agonists , Lipid Peroxides/metabolism , Mouth/metabolism , Mouth/surgery , Oxidation-Reduction/drug effects , Saliva/chemistry , Saliva/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Tumor Suppressor Protein p53/agonists , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
PLoS Pathog ; 11(9): e1005155, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26367131

ABSTRACT

Viral fulminant hepatitis (FH) is a severe disease with high mortality resulting from excessive inflammation in the infected liver. Clinical interventions have been inefficient due to the lack of knowledge for inflammatory pathogenesis in the virus-infected liver. We show that wild-type mice infected with murine hepatitis virus strain-3 (MHV-3), a model for viral FH, manifest with severe disease and high mortality in association with a significant elevation in IL-1ß expression in the serum and liver. Whereas, the viral infection in IL-1ß receptor-I deficient (IL-1R1-/-) or IL-1R antagonist (IL-1Ra) treated mice, show reductions in virus replication, disease progress and mortality. IL-1R1 deficiency appears to debilitate the virus-induced fibrinogen-like protein-2 (FGL2) production in macrophages and CD45+Gr-1high neutrophil infiltration in the liver. The quick release of reactive oxygen species (ROS) by the infected macrophages suggests a plausible viral initiation of NLRP3 inflammasome activation. Further experiments show that mice deficient of p47phox, a nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunit that controls acute ROS production, present with reductions in NLRP3 inflammasome activation and subsequent IL-1ß secretion during viral infection, which appears to be responsible for acquiring resilience to viral FH. Moreover, viral infected animals in deficiencies of NLRP3 and Caspase-1, two essential components of the inflammasome complex, also have reduced IL-1ß induction along with ameliorated hepatitis. Our results demonstrate that the ROS/NLRP3/IL-1ß axis institutes an essential signaling pathway, which is over activated and directly causes the severe liver disease during viral infection, which sheds light on development of efficient treatments for human viral FH and other severe inflammatory diseases.


Subject(s)
Carrier Proteins/agonists , Coronavirus Infections/virology , Host-Pathogen Interactions , Interleukin-1beta/agonists , Liver/virology , Murine hepatitis virus/physiology , Receptors, Interleukin-1 Type I/agonists , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Coronavirus Infections/immunology , Coronavirus Infections/metabolism , Coronavirus Infections/pathology , Disease Progression , Fibrinogen/metabolism , Immunity, Innate , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/ultrastructure , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Murine hepatitis virus/drug effects , Murine hepatitis virus/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , RAW 264.7 Cells , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Receptors, Interleukin-1 Type I/antagonists & inhibitors , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Survival Analysis
6.
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
7.
Chem Res Toxicol ; 29(3): 421-9, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-26901245

ABSTRACT

Our previous studies suggested that tetrachlorobenzoquinone (TCBQ) elicits pro-inflammatory activities; however, the mechanism of its toxicity toward vascular endothelial cell has not been characterized. Although TCBQ has been shown to stimulate interleukin-1 beta (IL-1ß) expression, it is unknown whether TCBQ regulates post-translational IL-1ß activation. Using human umbilical vein endothelial cells, we discovered that TCBQ not only promotes the expression of NOD-like receptor family, pyrin domain-containing protein 3 (NLRP3) components [composed of NLRP3, adaptor molecule apoptosis-associated speck like protein containing a caspase activation and recruitment domain (ASC), and pro-caspase 1] but also participates in priming the NLRP3 inflammasome. Activation of the NLRP3 inflammasome results in the maturation and release of IL-1ß. Further experiments showed that K(+) efflux, reactive oxygen species (ROS) production, and mitochondrial DNA damage may be involved in NLRP3 inflammasome activation mediated by TCBQ. Moreover, TCBQ downregulates the ubiquitination of NLRP3, further facilitating the activation of the NLRP3 inflammasome. These results suggest that the NLRP3/IL-1ß signaling pathway plays an important role in TCBQ-induced endothelial cell pro-inflammatory responses, which may point to potential therapeutic approaches against TCBQ-mediated toxicity.


Subject(s)
Benzoquinones/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Hydrocarbons, Chlorinated/pharmacology , Inflammasomes/drug effects , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Processing, Post-Translational/drug effects , Benzoquinones/chemistry , Cells, Cultured , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrocarbons, Chlorinated/chemistry , Inflammasomes/metabolism , Interleukin-1beta/agonists , Structure-Activity Relationship
8.
Arch Toxicol ; 90(11): 2779-2792, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26659566

ABSTRACT

Uremic toxins are considered a risk factor for cardiovascular disorders in kidney diseases, but it is not known whether, under inflammatory conditions, they affect adhesion molecule expression on endothelial cells, which may play a critical role in acute kidney injury (AKI). In the present study, in cardiovascular surgery-related AKI patients, who are known to have high plasma levels of the uremic toxin indoxyl sulfate (IS), plasma levels of IL-1ß were found to be positively correlated with plasma levels of the adhesion molecule E-selectin. In addition, high E-selectin and IL-1ß expression were seen in the kidney of ischemia/reperfusion mice in vivo. We also examined the effects of IS on E-selectin expression by IL-1ß-treated human umbilical vein endothelial cells (HUVECs) and the underlying mechanism. IS pretreatment of HUVECs significantly increased IL-1ß-induced E-selectin expression, monocyte adhesion, and the phosphorylation of mitogen-activated protein kinases (ERK, p38, and JNK) and transcription factors (NF-κB and AP-1), and phosphorylation was decreased by pretreatment with inhibitors of ERK1/2 (PD98059), p38 MAPK (SB202190), and JNK (SP600125). Furthermore, IS increased IL-1ß-induced reactive oxygen species (ROS) production and this effect was inhibited by pretreatment with N-acetylcysteine (a ROS scavenger) or apocynin (a NADPH oxidase inhibitor). Gel shift assays and ChIP-PCR demonstrated that IS enhanced E-selectin expression in IL-1-treated HUVECs by increasing NF-κB and AP-1 DNA-binding activities. Moreover, IS-enhanced E-selectin expression in IL-1ß-treated HUVECs was inhibited by Bay11-7082, a NF-κB inhibitor. Thus, IS may play an important role in the development of cardiovascular disorders in kidney diseases during inflammation by increasing endothelial expression of E-selectin.


Subject(s)
E-Selectin/metabolism , Endothelium, Vascular/drug effects , Indican/toxicity , Interleukin-1beta/agonists , MAP Kinase Signaling System/drug effects , Poisons/toxicity , Up-Regulation/drug effects , Acute Kidney Injury/blood , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Aged , Animals , Biomarkers/blood , Biomarkers/metabolism , Cells, Cultured , Disease Models, Animal , E-Selectin/chemistry , E-Selectin/genetics , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Human Umbilical Vein Endothelial Cells , Humans , Indican/blood , Interleukin-1beta/metabolism , Kidney/blood supply , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Male , Mice, Inbred C57BL , Oxidative Stress/drug effects , Poisons/blood , Reperfusion Injury/blood , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Uremia/etiology
9.
J Biol Chem ; 285(52): 40612-23, 2010 Dec 24.
Article in English | MEDLINE | ID: mdl-20956527

ABSTRACT

Smac mimetic compounds (SMCs) potentiate TNFα-mediated cancer cell death by targeting the inhibitor of apoptosis (IAP) proteins. In addition to TNFα, the tumor microenvironment is exposed to a number of pro-inflammatory cytokines, including IL-1ß. Here, we investigated the potential impact of IL-1ß on SMC-mediated death of cancer cells. Synergy was seen in a subset of a diverse panel of 21 cancer cell lines to the combination of SMC and IL-1ß treatment, which required IL-1ß-induced activation of the NF-κB pathway. Elevated NF-κB activity resulted in the production of TNFα, which led to apoptosis dependent on caspase-8 and RIP1. In addition, concurrent silencing of cIAP1, cIAP2, and X-linked IAP by siRNA was most effective for triggering IL-1ß-mediated cell death. Importantly, SMC-resistant cells that produced TNFα in response to IL-1ß treatment were converted to an SMC-sensitive phenotype by c-FLIP knockdown. Reciprocally, ectopic expression of c-FLIP blocked cell death caused by combined SMC and IL-1ß treatment in sensitive cancer cells. Together, our study indicates that a positive feed-forward loop by pro-inflammatory cytokines can be exploited by SMCs to induce apoptosis in cancer cells.


Subject(s)
Alkynes/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Dipeptides/pharmacology , Interleukin-1beta/pharmacology , Neoplasms/drug therapy , Peptidomimetics/pharmacology , Alkynes/agonists , Animals , Antineoplastic Agents/agonists , Baculoviral IAP Repeat-Containing 3 Protein , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cell Line, Tumor , Dipeptides/agonists , Drug Screening Assays, Antitumor , Drug Synergism , Gene Knockdown Techniques , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Interleukin-1beta/agonists , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Peptidomimetics/agonists , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases
10.
Eur J Immunol ; 39(9): 2617-28, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19676073

ABSTRACT

Cross-linking of CD137 ligand (CD137L), a member of the TNF family, with recombinant CD137-Fc (rCD137-Fc) protein enhanced adherence of bone marrow-derived macrophages, and increased the expression of ICAM-1, IL-1beta, IL-6, M-CSF and phosphotyrosine proteins. In RAW264.7 cells, a murine myeloid cell line, rCD137-Fc not only increased adherence but also cell multiplication, in a manner comparable to LPS or M-CSF. In addition, it up-regulated expression of IL-1beta, IL-1 receptor antagonist, IL-6, COX2, tenascin C, neuropeptide Y and M-CSF mRNA. Neutralization of M-CSF by incubating the RAW264.7 cells with anti-M-CSF mAb did not prevent the CD137L signal-induced viability. Viability was blocked by PP2, an Src tyrosine kinase inhibitor, rapamycin, an mTOR inhibitor and LY294002, a PI3K inhibitor, but not by Wortmannin, another PI3K inhibitor. Cross-linking of CD137L increased phosphorylation of Akt and p70S6 kinase. The latter was blocked by PP2, rapamycin or LY294002, but not by Wortmannin, whereas phosphorylation of Akt was blocked by LY294002 or Wortmannin. These findings demonstrate that reverse signals evoked by CD137L regulate immune functions in macrophages.


Subject(s)
4-1BB Ligand/metabolism , Carrier Proteins/immunology , Cell Survival/immunology , Macrophages/immunology , Phosphotransferases (Alcohol Group Acceptor)/immunology , Proto-Oncogene Proteins c-akt/immunology , 4-1BB Ligand/immunology , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Line , Cell Survival/drug effects , Chromones/pharmacology , Cytokines/drug effects , Cytokines/immunology , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Immunity, Innate , Immunosuppressive Agents/pharmacology , Intercellular Adhesion Molecule-1/drug effects , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1beta/agonists , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/agonists , Interleukin-6/immunology , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Macrophage Colony-Stimulating Factor/agonists , Macrophage Colony-Stimulating Factor/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Morpholines/pharmacology , Myeloid Cells/drug effects , Myeloid Cells/immunology , Phosphorylation/drug effects , Phosphorylation/immunology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tumor Necrosis Factor Receptor Superfamily, Member 9/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/immunology , src-Family Kinases/metabolism
11.
Immunology ; 128(1 Suppl): e532-40, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19740314

ABSTRACT

A new proinflammatory cytokine interleukin-32 (IL-32) has six isoforms. Although IL-32 can be detected in sera from patients suffering from Crohn's disease and rheumatoid arthritis, it is unclear which isoforms are involved. To this end, we investigated the functions of the most abundant IL-32beta by generating K562-IL-32beta stable cell lines. This report confirms, using IL-32 small interfering RNA, that IL-32beta induces an anti-inflammatory cytokine IL-10 in K562-IL-32beta cells and U937 promonocytic cells, which express endogenous IL-32beta upon phorbol 12-myristate 13-acetate (PMA) treatment, and monocyte-derived dendritic cells (DC) upon lipopolysaccharide (LPS) treatment. Interleukin-32beta was induced in monocyte-derived macrophages by LPS and in monocyte-derived DC by LPS, poly(I:C), or anti-CD40 antibody, but was not induced by PMA. We showed that IL-32beta expression was increased in a time-dependent manner in monocyte-derived DC upon LPS treatment and peaked at 24 hr. Production of IL-10 was exactly coincident with IL-32beta expression, but IL-1beta and tumour necrosis factor-alpha production peaked at 6 hr after LPS treatment, then steeply declined. Interleukin-12 p40 was induced at 9 hr and gradually increased until 48 hr, at which time IL-32beta and IL-10 were no longer increased. Knock-down of IL-32beta by IL-32 small interfering RNA led to the decrease of IL-10, but the increase of IL-12 in monocyte-derived DC, which means that IL-32beta promotes IL-10 production, but limits IL-12 production. We also showed that IL-10 neutralization increases IL-12, IL-1beta and tumour necrosis factor-alpha production, which implies that IL-10 suppresses such proinflammatory cytokines. Taken together, our results suggest that IL-32beta upregulates the production of an anti-inflammatory cytokine IL-10, and then IL-10 suppresses proinflammatory cytokines.


Subject(s)
Dendritic Cells/immunology , Interleukin-10/biosynthesis , Interleukins/metabolism , Macrophages/immunology , Cell Line, Tumor , Dendritic Cells/drug effects , Gene Knockdown Techniques , Humans , Interferon Inducers/pharmacology , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-1beta/agonists , Interleukin-1beta/metabolism , Interleukins/agonists , Interleukins/genetics , Lipopolysaccharides/immunology , Macrophages/drug effects , Poly I-C/pharmacology , Protein Isoforms/agonists , Protein Isoforms/immunology , RNA, Small Interfering/immunology , RNA, Small Interfering/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/metabolism
12.
J Clin Immunol ; 29(4): 508-16, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19291375

ABSTRACT

INTRODUCTION: Interleukin-1 beta (IL-1beta) and tumor necrosis factor alpha (TNFalpha) are potentially important in Chronic Obstructive Pulmonary Disease (COPD), but little is known of the relationships between these cytokines and their antagonists in disease compared with healthy controls. It is unclear if concentrations relate to disease severity. The study aimed to investigate these relationships and to assess the potential activity of each cytokine in the context of their antagonists. METHODS: Plasma cytokines, soluble receptors, and cell counts were measured in patients with stable COPD and age-matched healthy controls (n = 15 for both) daily for 5 days; these mediators were also measured in corresponding sputum samples from the COPD patients. RESULTS: COPD patients had significantly reduced concentrations of the antagonists, IL-1sRII, and IL-1RA compared with controls. In COPD, IL-1beta exceeded its antagonists and correlated significantly with BMI and FEV1, while plasma IL-1RA correlated positively with BMI but negatively with sputum IL-1beta, neutrophil, and macrophage counts and smoking history. TNFalpha antagonists exceeded agonists in both groups and did not correlate with COPD severity. CONCLUSIONS: Endogenous IL-1beta antagonists appear reduced in COPD. Furthermore, IL-1beta correlated with clinical aspects of disease severity, suggesting that IL-1beta may play a critical role in COPD. Given the relevant concentrations and binding affinities, it is likely that TNFalpha has limited activity in stable COPD.


Subject(s)
Cytokines/blood , Interleukin-1beta/blood , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/immunology , Tumor Necrosis Factor-alpha/blood , Aged , Aged, 80 and over , Female , Humans , Interleukin 1 Receptor Antagonist Protein/blood , Interleukin-1beta/agonists , Interleukin-1beta/antagonists & inhibitors , Male , Middle Aged , Sputum/immunology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/antagonists & inhibitors
13.
Blood Cells Mol Dis ; 43(3): 298-303, 2009.
Article in English | MEDLINE | ID: mdl-19766513

ABSTRACT

The purpose of this multidisciplinary investigation was to characterize cytokine production by human blood mononuclear cells after 2 contrasting exercise bouts (a maximal graded oxygen consumption [VO(2)max] test and 90 min of cycling at 85% of ventilatory threshold [VT]) when stimulated in vitro with extracts from bloodroot (Sanguinaria canadensis), coneflower (Echinacea tennesseensis), or solvent vehicle controls. Blood was sampled pre- and post-exercise. Production of TNF, IL-1beta, and IL-10 were measured at 24, 48, and 72 h, respectively. In the VO(2)max test there was a main effect of exercise such that exercise increased cytokine synthesis and a main effect of stimulant such that bloodroot extracts significantly increased cytokine production compared to other stimulants or controls. In the 90-min bout, there was a main effect of exercise for TNF and IL-1beta (but not IL-10) such that exercise decreased cytokine synthesis and a main effect of stimulant such that bloodroot extracts significantly increased cytokine production compared to other stimulants or controls, with exercisexstimulant interactions for both IL-1beta and IL-10. A similar though weaker effect was seen with Echinacea extracts; subsequent biochemical analyses suggested this was related to alkamide decay during 3 years undisturbed storage at ultralow (-80 degrees C) temperature. In this study, the VO(2)max test was associated with enhanced cytokine production whereas the 90-min cycling at 85% VT was associated with suppressed cytokine production. Bloodroot extracts were able to increase cytokine production in both contexts. Herbal extracts purported to offset exercise-associated effects on immune activity warrant continued investigation.


Subject(s)
Benzophenanthridines/pharmacology , Cytokines/blood , Echinacea , Exercise , Leukocytes, Mononuclear/drug effects , Sanguinaria , Adult , Amides/analysis , Amides/immunology , Amides/metabolism , Humans , Interleukin-10/agonists , Interleukin-10/blood , Interleukin-1beta/agonists , Interleukin-1beta/blood , Leukocytes, Mononuclear/immunology , Plant Extracts/pharmacology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/blood , Young Adult
14.
Neurochem Int ; 52(6): 1290-6, 2008 May.
Article in English | MEDLINE | ID: mdl-18328601

ABSTRACT

Amphotericin B (AmB) is a polyene antibiotic and reported to have therapeutic effects on prion diseases, in which the microglial activation has been suggested to play important roles by proliferating and producing various factors such as nitric oxide, proinflammatory cytokines, and so on. However, the therapeutic mechanism of AmB on prion diseases remains elusive. In the present study, we investigated the effects of AmB on cellular functions of rat primary cultured microglia. We found that AmB, similarly as lipopolysaccharide (LPS), could activate microglia to produce nitric oxide via inducible nitric oxide synthase. Both AmB and LPS also induced mRNA expressions of interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in microglia. AmB also changed the expression levels of neurotrophic factors mRNAs. AmB and LPS significantly down-regulated the level of ciliary neurotrophic factor mRNA. However, AmB, but not LPS, significantly up-regulated the level of glial cell-line derived neurotrophic factor mRNA in microglia. In addition, brain-derived neurotrophic factor mRNA expression level was tending upward by treatment with AmB, but not with LPS. Taken together, these results suggest that AmB regulates the microglial activation in different manner from LPS and that microglia may participate in the therapeutic effects of AmB on prion diseases by controlling the expression and production of such mediators.


Subject(s)
Amphotericin B/pharmacology , Gliosis/drug therapy , Inflammation/drug therapy , Microglia/drug effects , Prion Diseases/drug therapy , Amphotericin B/therapeutic use , Animals , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Brain-Derived Neurotrophic Factor/agonists , Brain-Derived Neurotrophic Factor/genetics , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cytokines/agonists , Cytokines/genetics , Glial Cell Line-Derived Neurotrophic Factor/agonists , Glial Cell Line-Derived Neurotrophic Factor/genetics , Gliosis/metabolism , Gliosis/physiopathology , Inflammation/metabolism , Inflammation/physiopathology , Interleukin-1beta/agonists , Interleukin-1beta/genetics , Interleukin-6/agonists , Interleukin-6/genetics , Microglia/metabolism , Nerve Growth Factors/agonists , Nerve Growth Factors/genetics , Neurotoxins/agonists , Neurotoxins/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Prion Diseases/metabolism , Prion Diseases/physiopathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
15.
Nutrients ; 10(6)2018 May 27.
Article in English | MEDLINE | ID: mdl-29861476

ABSTRACT

Fructose is a strong risk factor for non-alcoholic fatty liver disease (NAFLD), resulting from the disruption of redox systems by excessive reactive oxygen species production in the liver cells. Of note, recent epidemiological studies indicated that women are more prone to developing metabolic syndrome in response to fructose-sweetened beverages. Hence, we examined whether disruption of the redox system through a deletion of NADPH supplying mitochondrial enzyme, NADP⁺-dependent isocitrate dehydrogenase (IDH2), exacerbates fructose-induced NAFLD conditions in C57BL/6 female mice. Wild-type (WT) and IDH2 knockout (KO) mice were treated with either water or 34% fructose water over six weeks. NAFLD phenotypes and key proteins and mRNAs involved in the inflammatory pathway (e.g., NF-κB p65 and IL-1ß) were assessed. Hepatic lipid accumulation was significantly increased in IDH2 KO mice fed fructose compared to the WT counterpart. Neutrophil infiltration was observed only in IDH2 KO mice fed fructose. Furthermore, phosphorylation of NF-κB p65 and expression of IL-1ß was remarkably upregulated in IDH2 KO mice fed fructose, and expression of IκBα was decreased by fructose treatment in both WT and IDH2 KO groups. For the first time, we report our novel findings that IDH2 KO female mice may be more susceptible to fructose-induced NAFLD and the associated inflammatory response, suggesting a mechanistic role of IDH2 in metabolic diseases.


Subject(s)
Fatty Acids/metabolism , Fructose/adverse effects , Gene Expression Regulation , High Fructose Corn Syrup/adverse effects , Isocitrate Dehydrogenase/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Animals , Cytokines/blood , Female , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Interleukin-1beta/agonists , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Isocitrate Dehydrogenase/genetics , Liver/immunology , Liver/pathology , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Phosphorylation , Protein Processing, Post-Translational , Random Allocation , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
16.
Curr Stem Cell Res Ther ; 12(2): 175-180, 2017.
Article in English | MEDLINE | ID: mdl-26496886

ABSTRACT

The objective of this study was to investigate the effects of P. aeruginosa PAO1 cellular and soluble culture fractions on human mesenchymal stem cells (MSCs) death signaling pathways and cytokine profile. The bone marrow isolated MSCs, incubated for different periods of time with one of the three P. aeruginosa PAO1 culture fractions, i.e. low density whole cultures, heat inactivated bacterial cultures sediments and sterile supernatants, were submitted to the following assays: i) fluorescence microscopy evaluation of cellular morphology and viability; ii) bax, caspase 9, relA and bcl-2 genes expression analysis by qRT-PCR; and iii) quantification of the level of IL-1ß, IL-6, IL-8 and IL-10 cytokines released in the MSCs supernatants determined by ELISA. Results were statistically analyzed using the GraphPad In Stat software. The PAO1 whole cultures exhibited the most relevant influences, impacting on MSCs morphology and viability, interfering with apoptotic pathways and significantly stimulating the production of IL-1ß and IL-10, while decreasing the production of IL-6 and IL-8. The culture supernatants increased the production of IL-1ß and reduced the secretion of all other tested cytokines, while heat-inactivated bacterial cells significantly stimulated both IL-1ß and IL-10 production. These data could suggest that in vivo, the fate of P. aeruginosa infection depends on the proportion between different bacterial culture fractions (i.e. the number of viable bacterial cells, the number of dead cells and the amount of bacterial soluble products accumulated locally) that could be influenced by the initial infective dose, by the host defense mechanisms, and also by the administered antimicrobial treatment that may thus interfere with the evolution and magnitude of the induced lesions.


Subject(s)
Apoptosis/drug effects , Bone Marrow Cells/drug effects , Culture Media, Conditioned/pharmacology , Host-Pathogen Interactions , Mesenchymal Stem Cells/drug effects , Pseudomonas aeruginosa/chemistry , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Caspase 9/genetics , Caspase 9/immunology , Cell Survival/drug effects , Chemical Fractionation , Gene Expression Regulation , Hot Temperature , Humans , Interleukin-10/agonists , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-1beta/agonists , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-8/antagonists & inhibitors , Interleukin-8/genetics , Interleukin-8/immunology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/pathogenicity , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/immunology
17.
Biochem Pharmacol ; 140: 89-104, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28583844

ABSTRACT

Glioblastoma is among the most aggressive brain tumors and has an exceedingly poor prognosis. Recently, the importance of the tumor microenvironment in glioblastoma cell growth and progression has been emphasized. Toll-like receptor 4 (TLR4) recognizes bacterial lipopolysaccharide (LPS) and endogenous ligands originating from dying cells or the extracellular matrix involved in host defense and in inflammation. G-protein coupled receptors (GPCRs) have gained interest in anti-tumor drug discovery due to the role that they directly or indirectly play by transactivating other receptors, causing cell migration and proliferation. A proteomic analysis showed that the nociceptin receptor (NOPr) is among the GPCRs significantly expressed in glioblastoma cells, including U87 cells. We describe a novel role of the peptide nociceptin (N/OFQ), the endogenous ligand of the NOPr that counteracts cell migration, proliferation and increase in IL-1ß mRNA elicited by LPS via TLR4 in U87 glioblastoma cells. Signaling pathways through which N/OFQ inhibits LPS-mediated cell migration and elevation of [Ca2+]i require ß-arrestin 2 and are sensitive to TNFR-associated factor 6, c-Src and protein kinase C (PKC). LPS-induced cell proliferation and increase in IL-1ß mRNA are counteracted by N/OFQ via ß-arrestin 2, PKC and extracellular signal-regulated kinase 1/2; furthermore, the contributions of the transcription factors NF-kB and AP-1 were investigated. Independent of LPS, N/OFQ induces a significant increase in cell apoptosis. Contrary to what was observed in other cell models, a prolonged exposure to this endotoxin did not promote any tolerance of the cellular effects above described, including NOPr down-regulation while N/OFQ loses its inhibitory role.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Glioblastoma/drug therapy , Opioid Peptides/pharmacology , TNF Receptor-Associated Factor 6/agonists , Toll-Like Receptor 4/antagonists & inhibitors , beta-Arrestin 2/agonists , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/metabolism , Astrocytes/pathology , Calcium Signaling/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Glioblastoma/immunology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Interleukin-1beta/agonists , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins , Ligands , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , RNA Interference , Receptors, Opioid/agonists , Receptors, Opioid/genetics , TNF Receptor-Associated Factor 6/antagonists & inhibitors , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , beta-Arrestin 2/antagonists & inhibitors , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism , Nociceptin Receptor , Nociceptin
18.
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
19.
Neuropsychopharmacology ; 31(12): 2619-26, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16823390

ABSTRACT

The therapeutic use of interferon-alpha (IFN-alpha), a proinflammatory cytokine, is known to cause various neuropsychiatric adverse effects. In particular, depression occurs in 30-45% of patients, frequently interrupting treatment. IFN-alpha-treated animals also show depression-like behaviors. However, mechanisms underlying the depression caused by IFN-alpha remain to be defined. Recently, a decrease in adult hippocampal neurogenesis was revealed as a possible neuropathological mechanism of depression. Therefore, we investigated the effect of subchronic IFN-alpha treatment on neurogenesis in the adult rat dentate gyrus (DG). Immediately after the administration of IFN-alpha for 1 week, a decrease in the number of 5-bromo-deoxyuridine-labeled proliferating cells was observed in the DG; however, no effect was detected on the expression of mature neuronal phenotype in the newly formed cells 3 weeks later. Also, an increase in the level of interleukin-1beta (IL-1beta), a major proinflammatory cytokine, was observed in the hippocampus following the administration of IFN-alpha. Furthermore, coadministration of an IL-1 receptor antagonist completely blocked the IFN-alpha-induced suppression of the cell-proliferative activity in the DG. Our results indicate that IFN-alpha suppresses neurogenesis in the DG, and that IL-1beta plays an essential role in the suppression. The decreased cell proliferation caused by IFN-alpha-induced IL-1beta may be responsible, at least in part, for IFN-alpha-induced depression.


Subject(s)
Cell Proliferation/drug effects , Dentate Gyrus/drug effects , Interferon-alpha/adverse effects , Interleukin-1beta/agonists , Neurons/drug effects , Animals , Bromodeoxyuridine , Cell Division/drug effects , Cell Division/immunology , Dentate Gyrus/immunology , Dentate Gyrus/physiopathology , Depressive Disorder/chemically induced , Depressive Disorder/immunology , Depressive Disorder/physiopathology , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Immunologic Factors/adverse effects , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Male , Neurons/immunology , Rats , Rats, Wistar , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/immunology
20.
J Nutr Biochem ; 32: 39-45, 2016 06.
Article in English | MEDLINE | ID: mdl-27142735

ABSTRACT

Palmitic acid (C16:0) and TLR2 ligand induce, but docosahexaenoic acid (DHA) inhibits monocyte activation. C16:0 and TLR2 or TLR4 ligand induce certain ER stress markers; thus, we determined whether ER stress induced by these agonists is sufficient to induce monocyte activation, and whether the ER stress is inhibited by DHA which is known to inhibit C16:0- or ligand-induced TLR activation. Monocyte activation and ER stress were assessed by TLR/inflammasome-induced IL-1ß production, and phosphorylation of IRE-1 and eIF2 and expression of CHOP, respectively in THP-1 cells. TLR2 ligand Pam3CSK4 induced phosphorylation of eIF2, but not phosphorylation of IRE-1 and CHOP expression. LPS also induced phosphorylation of both IRE-1 and eIF2 but not CHOP expression suggesting that TLR2 or TLR4 ligand, or C16:0 induces different ER stress responses. C16:0-, Pam3CSK4-, or LPS-induced IL-1ß production was inhibited by 4-phenylbutyric acid, an inhibitor of ER stress suggesting that IL-1ß production induced by these agonists is partly mediated through ER stress. Among two ER stress-inducing molecules, thapsigargin but not tunicamycin led to the expression of pro-IL-1ß and secretion of IL-1ß. Thus, not all types of ER stress are sufficient to induce inflammasome-mediated IL-1ß secretion in monocytes. Although both C16:0 and thapsigargin-induced IL-1ß secretion was inhibited by DHA, only C16:0-mediated ER stress was responsive to DHA. These findings suggest that the anti-inflammatory effects of DHA are at least in part mediated through modulating ER homeostasis and that the propensity of ER stress can be differentially modulated by the types of dietary fat we consume.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Docosahexaenoic Acids/metabolism , Endoplasmic Reticulum Stress , Inflammasomes/metabolism , Monocytes/metabolism , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers/metabolism , Cell Line , Docosahexaenoic Acids/therapeutic use , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Humans , Immunomodulation , Inflammasomes/drug effects , Inflammasomes/immunology , Interleukin-1beta/agonists , Interleukin-1beta/metabolism , Ligands , Lipopeptides/pharmacology , Lipopolysaccharides/toxicity , Monocytes/drug effects , Monocytes/immunology , Palmitic Acid/adverse effects , Palmitic Acid/metabolism , Phenylbutyrates/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction/drug effects , Thapsigargin/pharmacology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL