Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
1.
Arterioscler Thromb Vasc Biol ; 37(5): 983-989, 2017 05.
Article in English | MEDLINE | ID: mdl-28302628

ABSTRACT

OBJECTIVE: To investigate the relationship between 3 markers of apoptosis, that is, FADD (Fas-associated death domain-containing protein), caspase-3, and caspase-8, and incidence of coronary events (CEs) in a population-based cohort study. APPROACH AND RESULTS: In vitro experiments were performed to assess the response of the apoptotic biomarkers after Fas stimulation of peripheral blood mononuclear cells. The experiments showed significantly increased releases of FADD, caspase-3, and caspase-8 after Fas stimulation. The relationship between FADD, caspase-3, and caspase-8, respectively, and incidence of CEs was studied in 4284 subjects from the population-based Malmö Diet and Cancer Study. Cox' proportional hazards regression was used to examine the association between the apoptotic biomarkers and incidence of CE over a mean follow-up of 19 years. A total of 381 individuals had CE during the follow-up. High FADD at baseline was significantly associated with incident CE. In the highest compared with the lowest quartile of FADD, the risk factor adjusted hazards ratio for CE was 1.82 (95% confidence interval, 1.35-2.46; P for trend <0.001). A significant association was also found between caspase-8 and CE; the hazards ratio (Q4 versus Q1) was 1.90 (95% confidence interval, 1.39-2.60; P for trend <0.001) after adjustment for risk factors. No association was found between caspase-3 and CEs. CONCLUSIONS: High levels of FADD and caspase-8, but not caspase-3, were associated with increased incidence of CE in subjects from the general population. The in vitro experiments support the view that these biomarkers could reflect activation of the extrinsic apoptotic pathway.


Subject(s)
Apoptosis , Caspase 3/blood , Caspase 8/blood , Coronary Disease/blood , Coronary Disease/epidemiology , Fas-Associated Death Domain Protein/blood , Biomarkers/blood , Cells, Cultured , Coronary Disease/diagnostic imaging , Coronary Disease/pathology , Female , Humans , Incidence , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/enzymology , Linear Models , Male , Middle Aged , Multivariate Analysis , Prognosis , Proportional Hazards Models , Risk Factors , Sweden/epidemiology , Time Factors , Up-Regulation , fas Receptor/pharmacology
2.
Mol Cell ; 64(2): 236-250, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27746017

ABSTRACT

Caspase-8 activation can be triggered by death receptor-mediated formation of the death-inducing signaling complex (DISC) and by the inflammasome adaptor ASC. Caspase-8 assembles with FADD at the DISC and with ASC at the inflammasome through its tandem death effector domain (tDED), which is regulated by the tDED-containing cellular inhibitor cFLIP and the viral inhibitor MC159. Here we present the caspase-8 tDED filament structure determined by cryoelectron microscopy. Extensive assembly interfaces not predicted by the previously proposed linear DED chain model were uncovered, and were further confirmed by structure-based mutagenesis in filament formation in vitro and Fas-induced apoptosis and ASC-mediated caspase-8 recruitment in cells. Structurally, the two DEDs in caspase-8 use quasi-equivalent contacts to enable assembly. Using the tDED filament structure as a template, structural analyses reveal the interaction surfaces between FADD and caspase-8 and the distinct mechanisms of regulation by cFLIP and MC159 through comingling and capping, respectively.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/chemistry , Caspase 8/chemistry , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Fas-Associated Death Domain Protein/chemistry , Viral Proteins/chemistry , Amino Acid Sequence , Apoptosis/drug effects , Binding Sites , CARD Signaling Adaptor Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cryoelectron Microscopy , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Death Effector Domain , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Gene Expression , Humans , Jurkat Cells , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Viral Proteins/genetics , Viral Proteins/metabolism , fas Receptor/pharmacology
3.
Cell Death Dis ; 7: e2209, 2016 04 28.
Article in English | MEDLINE | ID: mdl-27124583

ABSTRACT

Glioblastoma (GBM) is one of the most aggressive types of cancer with limited therapeutic options and unfavorable prognosis. Stemness and non-classical epithelial-to-mesenchymal transition (ncEMT) features underlie the switch from normal to neoplastic states as well as resistance of tumor clones to current therapies. Therefore, identification of ligand/receptor systems maintaining this privileged state is needed to devise efficient cancer therapies. In this study, we show that the expression of CD95 associates with stemness and EMT features in GBM tumors and cells and serves as a prognostic biomarker. CD95 expression increases in tumors and with tumor relapse as compared with non-tumor tissue. Recruitment of the activating PI3K subunit, p85, to CD95 death domain is required for maintenance of EMT-related transcripts. A combination of the current GBM therapy, temozolomide, with a CD95 inhibitor dramatically abrogates tumor sphere formation. This study molecularly dissects the role of CD95 in GBM cells and contributes the rational for CD95 inhibition as a GBM therapy.


Subject(s)
Brain Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Neoplasm Recurrence, Local/genetics , fas Receptor/genetics , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/classification , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Combinations , Drug Synergism , Glioblastoma/classification , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Immunoglobulin G/pharmacology , Neoplasm Recurrence, Local/classification , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Primary Cell Culture , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/pharmacology , Signal Transduction , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Survival Analysis , Temozolomide , fas Receptor/metabolism , fas Receptor/pharmacology
4.
Anticancer Drugs ; 26(7): 716-27, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25850884

ABSTRACT

Glioblastoma is a disease characterized by rapid invasive tumour growth. Studies on the proapoptotic CD95/CD95L signalling pathway recently suggested a significant contribution of CD95 signalling towards the high degree of motility in glioma cells. Apogenix has developed APG101, a clinical phase II compound designed to bind and neutralize CD95L, and thus to interfere with CD95/CD95L-based signalling. APG101 has shown clinical efficacy in a controlled randomized phase II trial in patients with recurrent glioma. Because APG101 is not cytotoxic to tumour cells in vitro, we postulated that the anti-invasive function of APG101 is the main mechanism of action for this compound. Using three-dimensional spheroid invasion assays in vitro and in murine brain tissue cultures, we found that knockdown of endogenous CD95L reduced the invasive phenotype in our two glioblastoma model cell lines U87-MG and U251-MG. Invasion was restored in CD95L knockdown cells upon the addition of soluble recombinant CD95L and this effect was inhibited by APG101. We conclude that CD95L from autocrine and paracrine sources contributes towards the invasive phenotype of glioblastoma cells and that APG101 acts as a suppressor of proinvasive signalling by the CD95/CD95L pathway in glioblastoma.


Subject(s)
Brain Neoplasms/pathology , Fas Ligand Protein/metabolism , Glioblastoma/pathology , Immunoglobulin G/pharmacology , Recombinant Fusion Proteins/pharmacology , Spheroids, Cellular/drug effects , fas Receptor/metabolism , Animals , Apoptosis/drug effects , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Drug Resistance, Neoplasm , Gene Knockdown Techniques , Glioblastoma/metabolism , Humans , Mice, Inbred C57BL , Neoplasm Invasiveness , Signal Transduction , Spheroids, Cellular/pathology , Spheroids, Cellular/physiology , fas Receptor/genetics , fas Receptor/pharmacology
5.
PLoS One ; 9(2): e90298, 2014.
Article in English | MEDLINE | ID: mdl-24587316

ABSTRACT

Eosinophils play a central role in asthma. The present study was performed to investigate the effect of tumour necrosis factor-α (TNF-α) on longevity of isolated human eosinophils. In contrast to Fas, TNF-α inhibited eosinophil apoptosis as evidenced by a combination of flow cytometry, DNA fragmentation assay and morphological analyses. The effect of TNF-α on eosinophil apoptosis was reversed by a TNF-α neutralising antibody. The anti-apoptotic effect of TNF-α was not due to autocrine release of known survival-prolonging cytokines interleukins 3 and 5 or granulocyte-macrophage-colony-stimulating factor as their neutralisation did not affect the effect of TNF-α. The anti-apoptotic signal was mediated mainly by the TNF-receptor 1. TNF-α induced phosphorylation and degradation of IκB and an increase in NF-κB DNA-binding activity. The survival-prolonging effect of TNF-α was reversed by inhibitors of NF-κB pyrrolidinedithiocarbamate and gliotoxin and by an inhibitor of IκB kinase, BMS-345541. TNF-α induced also an increase in AP-1 DNA-binding activity and the antiapoptotic effect of TNF-α was potentiated by inhibitors of AP-1, SR 11302 and tanshinone IIA and by an inhibitor of c-jun-N-terminal kinase, SP600125, which is an upstream kinase activating AP-1. Our results thus suggest that TNF-α delays human eosinophil apoptosis via TNF-receptor 1 and the resulting changes in longevity depend on yin-yang balance between activation of NF-κB and AP-1.


Subject(s)
Asthma/genetics , Eosinophils/metabolism , NF-kappa B/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Transcription Factor AP-1/genetics , Tumor Necrosis Factor-alpha/pharmacology , Antibodies/pharmacology , Apoptosis/drug effects , Asthma/metabolism , Asthma/pathology , Cell Survival/drug effects , Eosinophils/drug effects , Eosinophils/pathology , Gene Expression Regulation , Gliotoxin/pharmacology , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Imidazoles/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Phosphorylation , Primary Cell Culture , Protein Binding , Pyrrolidines/pharmacology , Quinoxalines/pharmacology , Receptors, Tumor Necrosis Factor, Type I/genetics , Retinoids/pharmacology , Signal Transduction , Thiocarbamates/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , fas Receptor/pharmacology
6.
Blood ; 121(3): 556-65, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23203823

ABSTRACT

Graft-versus-host disease (GVHD) induced by transplant-derived T cells represents a major complication after allogeneic bone marrow transplantation (BMT). However, these T cells support engraftment, early T-cell immunity, and mediate the graft-versus-tumor (GVT) effect. Cytotoxic effector functions by transplanted T cells are predominantly mediated by the perforin/granzyme and the CD95/CD95L system. APG101, a novel recombinant human fusion protein consisting of the extracellular domain of CD95 and the Fc domain of an IgG1 antibody inhibited CD95L-induced apoptosis without interfering with T-cell function in vitro and was therefore tested for its ability to prevent GVHD in murine BMT models across minor or major histocompatibility barriers. Starting APG101 treatment either 1 day before or 6 days after transplantation effectively reduced clinical GVHD and rescued survival between 60% and 100% if GVHD was CD95L mediated. APG101 did not interfere with the GVT effect, because P815 mastocytoma and most importantly primary Bcr-Abl-transformed B-cell leukemias were completely eradicated by the alloantigen-specific T cells. Phenotype and homing of alloantigen-specific T cells or their perforin/granzyme-mediated cytotoxicity and proliferative capacity were not affected by APG101 treatment suggesting that APG101 therapy might be useful in GVHD prophylaxis without impairing T-cell function and most importantly preserving GVT activity.


Subject(s)
Bone Marrow Transplantation/adverse effects , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Immunoglobulin G/pharmacology , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , fas Receptor/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Bone Marrow Transplantation/immunology , Cell Division/drug effects , Cell Division/immunology , Cell Movement/drug effects , Cell Movement/immunology , Disease Models, Animal , Female , Immunoglobulin G/immunology , Immunophenotyping , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasms/immunology , Recombinant Fusion Proteins/immunology , Transplantation, Homologous , fas Receptor/immunology
7.
J Neurotrauma ; 29(8): 1586-99, 2012 May 20.
Article in English | MEDLINE | ID: mdl-22260324

ABSTRACT

Spinal cord injury (SCI) is a devastating condition that currently lacks clinically-relevant and effective neuroprotective therapeutic options. Optimal therapeutic agents for clinical translation should show efficacy in a cervical compression/contusion model using a clinically-relevant post-injury therapeutic time window. To date, few compounds have met that rigorous standard. The objective of this work was to evaluate the efficacy of delayed post-injury administration of soluble Fas receptor (sFasR) via intrathecal catheter following acute cervical SCI in a clinically-relevant contusion/compression model. Female Wistar rats were given a C7-T1 moderately severe clip compression injury, followed by either 8-h or 24-h delayed treatment initiation. Long-term neurobehavioral analysis of motor recovery and neuropathic pain development was undertaken. The extent of oligodendrocyte and neuron survival was assessed in peri-lesional cord sections 8 weeks post-SCI. This was complemented by an evaluation of the level of tissue preservation at and adjacent to the site of injury. In animals treated with sFasR delayed 8 h post-injury, significant behavioral effects were observed, coinciding with enhanced cell survival, peri-lesional tissue sparing, and enhanced integrity of descending fiber tracts compared to control treatments. Animals treated with sFasR delayed by 24 h showed more modest improvements in behavioral recovery, and had consistent improvements in cell survival and tissue preservation. This work has shown for the first time that the Fas-mediated apoptotic pathway can be therapeutically targeted in a clinically-relevant time window post-SCI.


Subject(s)
Nerve Degeneration/drug therapy , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , fas Receptor/therapeutic use , Animals , Apoptosis/physiology , Behavior, Animal/drug effects , Female , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Neuralgia/drug therapy , Neuralgia/etiology , Neuralgia/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Pain Measurement , Rats , Rats, Wistar , Spinal Cord Injuries/complications , Spinal Cord Injuries/metabolism , Time Factors , fas Receptor/pharmacology
8.
PLoS One ; 6(5): e19974, 2011.
Article in English | MEDLINE | ID: mdl-21629700

ABSTRACT

Activation of an acid sphingomyelinase (aSMase) leading to a biosynthesis of GD3 disialoganglioside has been associated with Fas-induced apoptosis of lymphoid cells. The present study was undertaken to clarify the role of this enzyme in the generation of gangliosides during apoptosis triggered by Fas ligation. The issue was addressed by using aSMase-deficient and aSMase-corrected cell lines derived from Niemann-Pick disease (NPD) patients. Fas cross-linking elicited a rapid production of large amounts of complex a- and b-series species of gangliosides with a pattern and a chromatographic behavior as single bands reminiscent of brain gangliosides. The gangliosides were synthesized within the first ten minutes and completely disappeared within thirty minutes after stimulation. Noteworthy is the observation that GD3 was not the only ganglioside produced. The production of gangliosides and the onset of apoptotic hallmarks occurred similarly in both aSMase-deficient and aSMase-corrected NPD lymphoid cells, indicating that aSMase activation is not accountable for ganglioside generation. Hampering ganglioside production by inhibiting the key enzyme glucosylceramide synthase did not abrogate the apoptotic process. In addition, GM3 synthase-deficient lymphoid cells underwent Fas-induced apoptosis, suggesting that gangliosides are unlikely to play an indispensable role in transducing Fas-induced apoptosis of lymphoid cells.


Subject(s)
Apoptosis/drug effects , Gangliosides/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , fas Receptor/pharmacology , Blotting, Western , Cell Line , Cell Survival/drug effects , Flow Cytometry , Humans , Lymphocytes/drug effects , Sphingomyelins/metabolism
9.
Blood ; 117(22): 5953-62, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21478427

ABSTRACT

The most common form of neutrophil death is apoptosis. In the present study, we report surprising differences in the molecular mechanisms used for caspase activation between FAS/CD95-stimulated and TNF receptor 1 (TNFR1)-stimulated neutrophils. Whereas FAS-induced apoptosis was followed by caspase-8 activation and required Bid to initiate the mitochondrial amplification loop, TNF-α-induced apoptosis involved class IA PI3Ks, which were activated by MAPK p38. TNF-α-induced PI3K activation resulted in the generation of reactive oxygen species, which activated caspase-3, a mechanism that did not operate in neutrophils without active NADPH oxidase. We conclude that in neutrophils, proapoptotic pathways after TNFR1 stimulation are initiated by p38 and PI3K, but not by caspase-8, a finding that should be considered in anti-inflammatory drug-development strategies.


Subject(s)
Apoptosis , Neutrophils/pathology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Animals , BH3 Interacting Domain Death Agonist Protein/physiology , Caspase 3/metabolism , Caspase 8/metabolism , Cells, Cultured , Cytochromes c/metabolism , Humans , Membrane Potential, Mitochondrial , Mice , Mitochondria/metabolism , NADPH Oxidases/metabolism , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Biol Chem ; 285(26): 19997-20005, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20427266

ABSTRACT

Evidence accumulates that in clinically relevant cell death, both the intrinsic and extrinsic apoptotic pathway synergistically contribute to organ failure. In search for an inhibitor of apoptosis that provides effective blockage of these pathways, we analyzed viral proteins that evolved to protect the infected host cells. In particular, the cowpox virus protein crmA has been demonstrated to be capable of blocking key caspases of both pro-apoptotic pathways. To deliver crmA into eukaryotic cells, we fused the TAT protein transduction domain of HIV to the N terminus of crmA. In vitro, the TAT-crmA fusion protein was efficiently translocated into target cells and inhibited apoptosis mediated through caspase-8, caspase-9, and caspase-3 after stimulation with alpha-Fas, etoposide, doxorubicin, or staurosporine. The extrinsic apoptotic pathway was investigated following alpha-Fas stimulation. In vivo 90% of TAT-crmA-treated animals survived an otherwise lethal dose of alpha-Fas and showed protection from Fas-induced organ failure. To examine the intrinsic apoptotic pathway, we investigated the survival of mice treated with an otherwise lethal dose of doxorubicin. Whereas all control mice died within 31 days, 40% of mice that concomitantly received intraperitoneal injections of TAT-crmA survived. To test the ability to comprehensively block both the intrinsic and extrinsic apoptotic pathway in a clinically relevant setting, we employed a murine cardiac ischemia-reperfusion model. TAT-crmA reduced infarction size by 40% and preserved left ventricular function. In summary, these results provide a proof of principle for the inhibition of apoptosis with TAT-crmA, which might provide a new treatment option for ischemia-reperfusion injuries.


Subject(s)
Apoptosis/physiology , Recombinant Fusion Proteins/metabolism , Serpins/metabolism , Signal Transduction/physiology , Viral Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Caspase Inhibitors , Caspases/metabolism , Doxorubicin/pharmacology , Etoposide/pharmacology , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , Jurkat Cells , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Fluorescence , Myocardial Infarction/physiopathology , Myocardial Infarction/prevention & control , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Recombinant Fusion Proteins/pharmacology , Serpins/genetics , Serpins/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Survival Analysis , Viral Proteins/genetics , Viral Proteins/pharmacology , fas Receptor/pharmacology , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
11.
Endocrinology ; 151(2): 529-36, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19934374

ABSTRACT

Senescence marker protein-30 (SMP30) is an androgen-independent factor that decreases with age. We recently identified SMP30 as the lactone-hydrolyzing enzyme gluconolactonase (GNL), which is involved in vitamin C biosynthesis in animal species. To examine whether the age-related decrease in SMP30/GNL has effects on glucose homeostasis, we used SMP30/GNL knockout (KO) mice treated with L-ascorbic acid. In an ip glucose tolerance test at 15 wk of age, blood glucose levels in SMP30/GNL KO mice were significantly increased by 25% at 30 min after glucose administration compared with wild-type (WT) mice. Insulin levels in SMP30/GNL KO mice were significantly decreased by 37% at 30 min after glucose compared with WT mice. Interestingly, an insulin tolerance test showed a greater glucose-lowering effect in SMP30/GNL KO mice. High-fat diet feeding severely worsened glucose tolerance in both WT and SMP30/GNL KO mice. Morphometric analysis revealed no differences in the degree of high-fat diet-induced compensatory increase in beta-cell mass and proliferation. In the static incubation study of islets, insulin secretion in response to 20 mm glucose or KCl was significantly decreased in SMP30/GNL KO mice. On the other hand, islet ATP content at 20 mm in SMP30/GNL KO mice was similar to that in WT mice. Collectively, these data indicate that impairment of the early phase of insulin secretion due to dysfunction of the distal portion of the secretion pathway underlies glucose intolerance in SMP30/GNL KO mice. Decreased SMP30/GNL may contribute to the worsening of glucose tolerance that occurs in normal aging.


Subject(s)
Calcium-Binding Proteins/deficiency , Carboxylic Ester Hydrolases/deficiency , Carboxylic Ester Hydrolases/genetics , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Adipose Tissue/drug effects , Adipose Tissue/physiology , Aging/genetics , Aging/physiology , Animal Feed , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Brain/physiology , Cell Division/drug effects , Epididymis/anatomy & histology , Glucose Tolerance Test , Insulin/analysis , Insulin/pharmacology , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Islets of Langerhans/physiology , Liver/cytology , Liver/drug effects , Liver/physiology , Lung/physiology , Male , Mice , Mice, Knockout , Pulmonary Alveoli/pathology , Tumor Necrosis Factor-alpha/pharmacology , Weight Gain , fas Receptor/pharmacology
12.
Ren Fail ; 31(5): 377-81, 2009.
Article in English | MEDLINE | ID: mdl-19839838

ABSTRACT

UNLABELLED: Fas (APO-1/CD95) is a cell surface receptor that initiates apoptotic pathway. Fas-stimulated ROS generation may play important role in Fas-mediated apoptosis. The aim of this study was to evaluate the influence of interferon-alpha on oxidative stress parameters in Fas-induced renal apoptosis in mice kidney. SUBJECTS AND METHODS: One-month-old Balb C male mice were used for the study. The animals were divided in four groups: group 1 were the controls, group 2 mice were treated with anti-Fas antibody i.p., group 3 mice were treated with IFN-alpha, and group 4 mice were treated with both agents simultaneously. The mice were killed 48 h afterwards, and kidneys were homogenized. TBA reactive substances (TBARS), glutathione content, and reactive carbonyl group (RCG) were measured. RESULTS: The results showed a statistically significant increase of TBARS (p < 0.05) and RCG (p < 0.05) concentration in the group treated with anti-Fas antibody versus control. IFN-alpha decreased the concentration of TBARS and RCG after anti-Fas antibody administration (p < 0.05). There is no significant difference in glutathione content between investigated groups. CONCLUSION: IFN-alpha might be considered as a new target for therapeutic intervention in FasL/Fas induced renal injury.


Subject(s)
Apoptosis/drug effects , Interferon-alpha/pharmacology , Kidney/drug effects , fas Receptor/pharmacology , Animals , Apoptosis/physiology , Creatinine/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Kidney/metabolism , Male , Mice , Mice, Inbred BALB C , Oxidative Stress/drug effects , Probability , Random Allocation , Reference Values , Statistics, Nonparametric , Urea/blood
13.
Exp Neurol ; 215(1): 48-52, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18950622

ABSTRACT

Estrogen is protective in experimental cerebral ischemia, yet the mechanism remains unclear. Fas-mediated apoptosis has been shown to be induced after cerebral ischemia and significantly contribute to ischemic brain damage. In this study, we tested if estrogen is protective against cerebral ischemia by suppressing Fas-mediated apoptosis. 17Beta-estradiol-treated and untreated ovariectomized (OVX) female mice were subjected to 2 h middle cerebral artery occlusion (MCAO). Expression of Fas and Fas-associated death domain (FADD) were measured at 3, 6 and 12 h of reperfusion by RT-PCR and Western blot, respectively. Post-ischemic activities of caspase-8 and -3 activities, the two downstream effectors of Fas-induced apoptosis, were also assayed at same time points by ELISA. Finally, Fas antibody-induced cell death in primary cortical neurons was assayed by fluorescence activated cell sorter (FACS) in the presence and absence of estradiol. Our data showed that estradiol-treated OVX female mice sustained smaller infarct compared to untreated OVX mice. Ischemia upregulated Fas and FADD expression, and increased caspase-8 and -3 activities in OVX female mouse cortex, which were significantly attenuated by estradiol. Estradiol also significantly inhibited Fas antibody-induced neuronal cell apoptosis. Our data suggests that inhibition of ischemia-induced Fas-mediated apoptosis is an important mechanism of neuroprotection by estrogen in cerebral ischemia.


Subject(s)
Apoptosis/drug effects , Brain Infarction/prevention & control , Estradiol/therapeutic use , Infarction, Middle Cerebral Artery/complications , fas Receptor/pharmacology , Analysis of Variance , Animals , Brain Infarction/etiology , Caspase 3/metabolism , Caspase 8/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Embryo, Mammalian , Estradiol/pharmacology , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Female , Flow Cytometry/methods , Gene Expression Regulation/drug effects , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/therapy , Neurons/drug effects , Ovariectomy , Rats
14.
Cancer Immunol Immunother ; 58(5): 699-708, 2009 May.
Article in English | MEDLINE | ID: mdl-18807035

ABSTRACT

Several tumor immunotherapy approaches result in a low percentage of durable responses in selected cancers. We hypothesized that the insensitivity of cancer cells to immunotherapy may be related to an anti-apoptotic cancer cell milieu, which could be pharmacologically reverted through the inhibition of antiapoptotic Bcl-2 family proteins in cancer cells. ABT-737, a small molecule inhibitor of the antiapoptotic proteins Bcl-2, Bcl-w and Bcl-x(L), was tested for the ability to increase antitumor immune responses in two tumor immunotherapy animal models. The addition of systemic therapy with ABT-737 to the immunization of BALB/c mice with tumor antigen peptide-pulsed dendritic cells (DC) resulted in a significant delay in CT26 murine colon carcinoma tumor growth and improvement in survival. However, the addition of ABT-737 to either a vaccine strategy involving priming with TRP-2 melanoma antigen peptide-pulsed DC and boosting with recombinant Listeria monocytogenes expressing the same melanoma antigen, or the adoptive transfer of TCR transgenic cells, did not result in superior antitumor activity against B16 murine melanoma. In vitro studies failed to demonstrate increased cytotoxic lytic activity when testing the combination of ABT-737 with lymphokine activated killer (LAK) cells, or the death receptor agonists Fas, TRAIL-ligand or TNF-alpha against the CT26 and B16 cell lines. In conclusion, the Bcl-2 inhibitor ABT-737 sensitized cancer cells to the antitumor effect of antigen-specific immunotherapy in a vaccine model for the CT26 colon carcinoma in vivo but not in two immunotherapy strategies against B16 melanoma.


Subject(s)
Biphenyl Compounds/therapeutic use , Cancer Vaccines/therapeutic use , Colonic Neoplasms/therapy , Immunotherapy/methods , Melanoma, Experimental/therapy , Neoplasm Proteins/antagonists & inhibitors , Nitrophenols/therapeutic use , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/therapeutic use , Animals , Antigens, Neoplasm/immunology , Apoptosis/drug effects , Apoptosis/immunology , Cancer Vaccines/immunology , Cell Line, Tumor/drug effects , Cell Line, Tumor/immunology , Colonic Neoplasms/immunology , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Drug Screening Assays, Antitumor , Humans , Immunotherapy, Adoptive , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Killer Cells, Lymphokine-Activated/transplantation , Listeria monocytogenes/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Piperazines/therapeutic use , Receptors, Death Domain/agonists , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/pharmacology
15.
Anticancer Res ; 28(2A): 593-9, 2008.
Article in English | MEDLINE | ID: mdl-18506997

ABSTRACT

BACKGROUND: Acquired resistance to apoptosis is a critical event in tumour development and in insensitivity toward therapy. To investigate resistance mechanisms to Fas/CD95/Apo-1-induced apoptosis, a Fas ligand-resistant variant of the U937 cell line was generated. RESULTS: Selection for Fas resistance resulted in a partial cross-resistance to TRAIL and TNF-alpha. Activation of caspase-8 was found to be impaired and the expression of Fas was reduced. However, FADD expression and ligand-induced aggregation of Fas was intact. Inhibition of various signalling pathways with pharmacological inhibitors revealed that resistance to death receptor-mediated apoptosis was dependent on altered tyrosine phosphatase/kinase activities and de novo protein synthesis. Moreover, FLIP, an anti-apoptotic protein, was expressed to a higher extent in the resistant cells. CONCLUSION: We provide evidence that acquired resistance to Fas-induced apoptosis in U937 cells involves a discrete set of molecular mechanisms which also render the cells cross-resistant to other death ligands.


Subject(s)
Caspase 8/metabolism , Drug Resistance, Neoplasm , fas Receptor/pharmacology , Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Enzyme Activation , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/pharmacology , Humans , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
16.
Oncol Rep ; 19(6): 1577-82, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18497968

ABSTRACT

A-kinase-anchoring protein 149 (AKAP149) is a member of a structurally diverse, though functionally similar anchoring protein family and is localized to the outer membrane of mitochondria and in the endoplasmic reticulum-nuclear envelope network. AKAP149 plays an important role in controlling the subcellular localization and temporal specificity of protein phosphorylation and mRNA metabolism by tethering kinases and phosphatases, such as protein kinase A and type I protein phosphatase, through its N-terminal protein-binding motifs and mRNAs via its C-terminal RNA-binding motifs. It is well recognized that caspases play a central role in transducing and amplifying the intracellular death signal and that apoptosis is executed as a consequence of caspase-mediated cleavage of multiple cellular substrates. The identification of novel death substrates and elucidation of the consequences of their proteolytic cleavages by caspases are therefore crucial for our understanding of cell death and other biological processes. Herein, we demonstrated that AKAP149 is a direct substrate of active caspase-3, -8 -and -10 in vitro and in vivo. 35S-labeled full-length AKAP149 was completely cleaved in vitro by active caspase-3, -8 and -10 into two fragments of approximately 105 and 45 kDa, while caspase-2 cleaved it partially and caspase-1 did not cleave it at all. AKAP149 was also cleaved by caspases during Fas- and staurosporine-induced apoptosis in Jurkat T and HeLa cells, which were blocked by specific inhibitors of caspase-3 and -8. The specific cleavage site for these caspases was mapped in vitro and in vivo to Asp582 at AKAP149, which is located between the protein kinase A regulatory subunit anchoring and KH RNA-binding domains. In addition, HeLa cells transiently overexpressing AKAP149 D582E mutant were resistant to staurosporine-induced HeLa cell apoptosis. Taken together, these data suggest that AKAP149 activity may be deregulated by caspase-dependent proteolysis during apoptotic cell death and may provide useful information for elucidating the apoptosis signaling pathways in detail.


Subject(s)
A Kinase Anchor Proteins/metabolism , Apoptosis/physiology , Caspases/metabolism , A Kinase Anchor Proteins/genetics , Aspartic Acid/chemistry , Aspartic Acid/genetics , HeLa Cells , Humans , Jurkat Cells , Mutation/genetics , Staurosporine/pharmacology , fas Receptor/pharmacology
17.
Oncogene ; 27(18): 2513-24, 2008 Apr 17.
Article in English | MEDLINE | ID: mdl-17982483

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has emerged as a promising antineoplastic agent because of its ability to selectively kill tumoral cells. However, some cancer cells are resistant to TRAIL-induced apoptosis. We have previously demonstrated that in endometrial carcinoma cells such resistance is caused by elevated FLICE-inhibitory protein (FLIP) levels. The present study focuses on the mechanisms by which FLIP could be modulated to sensitize endometrial carcinoma cells to TRAIL-induced apoptosis. We find that inhibition of casein kinase (CK2) sensitizes endometrial carcinoma cells to TRAIL- and Fas-induced apoptosis. CK2 inhibition correlates with a reduction of FLIP protein, suggesting that CK2 regulates resistance to TRAIL and Fas by controlling FLIP levels. FLIP downregulation correlates with a reduction of mRNA and is prevented by addition of the MG-132, suggesting that CK2 inhibition results in a proteasome-mediated degradation of FLIP. Consistently, forced expression of FLIP restores resistance to TRAIL and Fas. Moreover, knockdown of either FADD or caspase-8 abrogates apoptosis triggered by inhibition of CK2, indicating that CK2 sensitization requires formation of functional DISC. Finally, because of the possible role of both TRAIL and CK2 in cancer therapy, we demonstrate that CK2 inhibition sensitizes primary endometrial carcinoma explants to TRAIL apoptosis. In conclusion, we demonstrate that CK2 regulates endometrial carcinoma cell sensitivity to TRAIL and Fas by regulating FLIP levels.


Subject(s)
Antineoplastic Agents/pharmacology , CASP8 and FADD-Like Apoptosis Regulating Protein/biosynthesis , Casein Kinase II/metabolism , Drug Resistance, Neoplasm/drug effects , Endometrial Neoplasms/metabolism , Neoplasm Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , fas Receptor/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Casein Kinase II/antagonists & inhibitors , Cell Line , Drug Resistance, Neoplasm/genetics , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Leupeptins/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Tumor Cells, Cultured , fas Receptor/therapeutic use
18.
Am J Pathol ; 171(6): 1831-46, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17991716

ABSTRACT

Hepatitis C virus (HCV) is a major cause of chronic hepatitis that can lead to cirrhosis and hepatocellular carcinoma. To study the effects of HCV protein expression on host cells, we established conditional expression of the full-length open reading frame (ORF) of an infectious cDNA clone of HCV (genotype 1a, H77 strain) in the nontransformed human hepatocyte line cell HH4 using the ecdysone receptor regulatory system. Treatment with the ecdysone analog ponasterone-A induced tightly regulated and dose-dependent full-length HCV ORF expression and properly processed HCV proteins. HCV Core, NS3, and NS5A colocalized in perinuclear regions and associated with the early endosomal protein EEA1. HCV ORF expression caused marked growth inhibition, increased intracellular reactive oxygen species, up-regulation of glutamate-l-cysteine ligase activity, increased glutathione level, and activation of nuclear factor kappaB. Although it was not directly cytotoxic, HCV ORF expression sensitized HH4 cells to Fas at certain concentrations but not to tumor necrosis factor-related apoptosis-inducing ligand. HCV ORF expression in HH4 cells up-regulated genes involved in innate immune response/inflammation and oxidative stress responses and down-regulated cell growth-related genes. Expression of HCV ORF in host cells may contribute to HCV pathogenesis by producing oxidative stress and increasing the expression of genes related to the innate immune response and inflammation.


Subject(s)
Hepacivirus/metabolism , Hepatocytes/immunology , Hepatocytes/virology , Immunity, Innate/genetics , Viral Proteins/metabolism , Apoptosis/genetics , Cell Adhesion/genetics , Cell Cycle/genetics , Cell Proliferation , Cell Survival/genetics , Ecdysterone/analogs & derivatives , Ecdysterone/pharmacology , Extracellular Matrix/genetics , Gene Expression , Glutamate-Cysteine Ligase/metabolism , Hepacivirus/genetics , Hepatocytes/chemistry , Humans , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Open Reading Frames/drug effects , Open Reading Frames/genetics , Oxidative Stress/genetics , Polyproteins/genetics , Polyproteins/metabolism , Transcription, Genetic , Up-Regulation , Viral Proteins/analysis , Viral Proteins/genetics , fas Receptor/pharmacology
19.
Exp Mol Med ; 39(4): 439-49, 2007 Aug 31.
Article in English | MEDLINE | ID: mdl-17934331

ABSTRACT

Neutrophils are also known to acquire the characteristics of dendritic cells (DCs) under the appropriate conditions. In this study, neutrophils were cultivated in vitro in the presence or absence of compounds modulating their survival in an attempt to characterize the expression profile of the DC markers. Higher MHC-II, CD80, CD86, CD83, and CD40 expression levels were detected on the surface of the cultured neutrophils for 24 h than on the freshly isolated cells. The annexin V-positive cells showed a higher expression level of the DC markers than the annexin V-negative cells. The population of neutrophils double stained with annexin V and the DC markers increased after being incubated with agonistic anti-Fas Ab. LPS, the anti-apoptotic compound, decreased the CD86 and MHC-II expression levels but 50-60% of the DC marker-positive cells were detected in the annexin V-positive cells. In contrast, CD80, CD86, CD83, and HLA-DR mRNA levels increased in the GM-CSF-treated neutrophils but not in the anti-Fas Ab-treated neutrophils. T cell proliferation was inhibited by co-culturing them with anti-Fas Ab- or LPS-treated neutrophils at a high neutrophil:T cell ratio. However, the superantigen-mediated T cell proliferation was increased by the LPS-treated neutrophils but decreased by the anti-Fas Ab-treated neutrophils. There was a lower level of interferon-gamma production in the T cells co-cultured with anti-Fas Ab-treated neutrophils than with the LPS-treated neutrophils. This suggests that apoptotic neutrophils express DC markers on their surface and the differential expression of DC markers might have a detrimental effect on the immune reaction.


Subject(s)
Antigens, Differentiation/biosynthesis , Apoptosis , Dendritic Cells/metabolism , Neutrophils/metabolism , Antigen Presentation , Antigens, CD/biosynthesis , Cells, Cultured , Humans , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Neutrophils/physiology , T-Lymphocytes/immunology , fas Receptor/pharmacology
20.
J Pharmacol Exp Ther ; 323(1): 31-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17620456

ABSTRACT

Simendans are novel agents used in the treatment of decompensated heart failure. They sensitize troponin C to calcium and open ATP-sensitive potassium channels and have been shown to reduce cardiac myocyte apoptosis. The aim of the present study was to evaluate whether simendans reduce pulmonary eosinophilia and regulate eosinophil apoptosis. Bronchoalveolar lavage (BAL) eosinophilia was evaluated in ovalbumin-sensitized mice. Effects of simendans on apoptosis in isolated human eosinophils were assessed by relative DNA fragmentation assay, annexin V-binding, and morphological analysis. Dextrosimendan [(+)-[[4-(1,4,5,6-tetrahydro-4-methyl-6-oxo-3-pyridazinyl)phenyl)hydrazono]propanedinitrile] reduced ovalbumin-induced BAL-eosinophilia in sensitized mice. Levosimendan [(-)-[[4-(1,4,5,6-tetrahydro-4-methyl-6-oxo-3-pyridazinyl)phenyl]hydrazono]propanedinitrile] and dextrosimendan reversed interleukin (IL)-5-afforded survival of human eosinophils by inducing apoptosis in vitro. Even high concentrations of IL-5 were not able to overcome the effect of dextrosimendan. Dextrosimendan further enhanced spontaneous apoptosis as well as that induced by CD95 ligation, without inducing primary necrosis. Dextrosimendan-induced DNA fragmentation was shown to be dependent on caspase and c-Jun NH2-terminal kinase activation, whereas extracellular signal-regulated kinase, p38 mitogen-activated kinase, and ATP-sensitive potassium channels seemed to play no role in its actions. Taken together, our results show that simendans possess antieosinophilic activity and may be useful for the treatment of eosinophilic inflammation.


Subject(s)
Apoptosis/drug effects , Eosinophils/drug effects , Hydrazones , Pulmonary Eosinophilia/drug therapy , Pyridazines , Animals , Bronchoalveolar Lavage Fluid/cytology , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , DNA Fragmentation/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Eosinophils/immunology , Humans , Hydrazones/pharmacology , Hydrazones/therapeutic use , Interleukin-5/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred BALB C , Ovalbumin/pharmacology , Potassium Channels/metabolism , Pulmonary Eosinophilia/immunology , Pyridazines/pharmacology , Pyridazines/therapeutic use , Simendan , fas Receptor/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...