Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
J Biochem Mol Toxicol ; 32(11): e22213, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30152906

ABSTRACT

We investigated the effect of apigenin, a dietary flavonoid, on isoproterenol hydrochloride (ISO)-induced apoptotic signaling in cardiomyoblast H9C2 cells. The results showed that apigenin treatment (10 µM) prevented ISO (31.25 µM)-induced lipid peroxidative levels and antioxidants status in H9C2 cells. Furthermore, apigenin inhibited expression of inflammatory markers in ISO-treated cells. In addition, apigenin prevented ISO-induced DNA damage and apoptotic signaling through modulating the expression of Bax, caspase-3, -8 and -9, cytochrome c, and Fas proteins in H9C2 cells. It is concluded that apigenin prevents ISO-induced antioxidants depletion, oxidative DNA damage, inflammatory, and apoptotic signaling in H9C2 cells. Thus, the present results demonstrated that apigenin has a cardioprotective effect on cardiomyoblasts cells.


Subject(s)
Antioxidants/pharmacology , Apigenin/pharmacology , Apoptosis/drug effects , Cardiotonic Agents/adverse effects , Isoproterenol/adverse effects , Myoblasts, Cardiac/drug effects , Oxidative Stress/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biomarkers/metabolism , Cardiotonic Agents/antagonists & inhibitors , Cell Line , DNA Damage/drug effects , Inflammation Mediators/agonists , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Isoproterenol/antagonists & inhibitors , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial , Mitochondria, Heart/drug effects , Mitochondria, Heart/immunology , Mitochondria, Heart/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/immunology , Mitochondrial Membranes/metabolism , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/immunology , Myoblasts, Cardiac/metabolism , Rats , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/metabolism , fas Receptor/agonists , fas Receptor/antagonists & inhibitors , fas Receptor/metabolism
2.
Diabetes Obes Metab ; 19(5): 682-694, 2017 05.
Article in English | MEDLINE | ID: mdl-28058779

ABSTRACT

AIMS: Islet amyloid, formed by aggregation of human islet amyloid polypeptide (hIAPP), contributes to ß-cell failure in type 2 diabetes, cultured and transplanted islets. We previously showed that biosynthetic hIAPP aggregates induce ß-cell Fas upregulation and activation of the Fas apoptotic pathway. We used cultured human and hIAPP-expressing mouse islets to investigate: (1) the role of interleukin-1ß (IL-1ß) in amyloid-induced Fas upregulation; and (2) the effects of IL-1ß-induced ß-cell dysfunction on pro-islet amyloid polypeptide (proIAPP) processing and amyloid formation. RESEARCH DESIGN AND METHODS: Human and h IAPP -expressing mouse islets were cultured to form amyloid without or with the IL-1 receptor antagonist (IL-1Ra) anakinra, in the presence or absence of recombinant IL-1ß. Human islets in which amyloid formation was prevented (amyloid inhibitor or Ad-prohIAPP-siRNA) were cultured similarly. ß-cell function, apoptosis, Fas expression, caspase-8 activation, islet IL-1ß, ß-cell area, ß-/α-cell ratio, amyloid formation, and (pro)IAPP forms were assessed. RESULTS: hIAPP aggregates were found to increase IL-1ß levels in cultured human islets that correlated with ß-cell Fas upregulation, caspase-8 activation and apoptosis, all of which were reduced by IL-1Ra treatment or prevention of amyloid formation. Moreover, IL-1Ra improved culture-induced ß-cell dysfunction and restored impaired proIAPP processing, leading to lower amyloid formation. IL-1ß treatment potentiated impaired proIAPP processing and increased amyloid formation in cultured human and h IAPP -expressing mouse islets, which were prevented by IL-1Ra. CONCLUSIONS: IL-1ß plays a dual role by: (1) mediating amyloid-induced Fas upregulation and ß-cell apoptosis; (2) inducing impaired proIAPP processing thereby potentiating amyloid formation. Blocking IL-1ß may provide a new strategy to preserve ß cells in conditions associated with islet amyloid formation.


Subject(s)
Amyloid/agonists , Apoptosis , Interleukin-1beta/metabolism , Islet Amyloid Polypeptide/metabolism , Islets of Langerhans/metabolism , fas Receptor/agonists , Adult , Amyloid/antagonists & inhibitors , Amyloid/chemistry , Amyloid/metabolism , Animals , Cadaver , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/surgery , Hemizygote , Humans , Insulin/metabolism , Insulin Secretion , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Islet Amyloid Polypeptide/antagonists & inhibitors , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/genetics , Islets of Langerhans/cytology , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/adverse effects , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Protein Precursors/antagonists & inhibitors , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Processing, Post-Translational , RNA Interference , Recombinant Proteins/metabolism , Tissue Culture Techniques , fas Receptor/metabolism
3.
Cell Physiol Biochem ; 39(4): 1433-43, 2016.
Article in English | MEDLINE | ID: mdl-27606894

ABSTRACT

BACKGROUND/AIMS: The hepatitis B virus X protein (HBx) contributes to HBV-induced injury of renal tubular cells and induces apoptosis via Fas/FasL up-regulation. However, the mechanism of Fas/FasL activation is unknown. Recent studies indicated that HBx induction of apoptosis in hepatic cells depends on activating the MLK3-MKK7-JNKs signaling module, which then up-regulates FasL expression. In this study, we used NRK-52E cells transfected an HBx expression vector to examine the role of the MLK3-MKK7-JNKs signaling pathway on HBx-induced renal tubular cell injury. METHODS: NRK-52E cells were transfected with pc-DNA3.1(+)-HBx to establish an HBx over-expression model, and with pc-DNA3.1(+)-HBx and pSilencer3.1-shHBx to establish an HBx low expression model. One control group was not transfected and another control group was transfected with an empty plasmid. Cell proliferation was determined by the formazan dye method (Cell Counting Kit-8) and apoptosis was measured by flow cytometry and fluorescence microscopy. Western blotting was used to measure the expression of Fas, FasL, and MLK3-MKK7-JNKs signaling pathway-related proteins. The activity of caspase-8 was measured by spectrophotometry. RESULTS: Transfection of NRK-52E cells with pc-DNA3.1(+)-HBx inhibited cell proliferation and increased apoptosis and caspase-8 activity. The expression of Fas, FasL, and MLK3-MKK7-JNKs signaling pathway-related proteins were also greater in the pc-DNA3.1(+)-HBx group, but lower in RNAi group. Furthermore, the activity of MLK3-MKK7-JNKs signaling pathway, expression of Fas/FasL, and apoptosis were significantly lower in the pc-DNA3.1(+)-HBx group when treated with K252a, a known inhibitor of MLK3. CONCLUSIONS: Our results show that HBx induces apoptosis in NRK-52E cells and activates Fas/FasL via the MLK3-MKK7-JNK3-c-Jun signaling pathway.


Subject(s)
Epithelial Cells/drug effects , Fas Ligand Protein/agonists , Hepatitis B virus/chemistry , Signal Transduction/genetics , Trans-Activators/pharmacology , fas Receptor/agonists , Animals , Apoptosis/drug effects , Carbazoles/pharmacology , Caspase 8/genetics , Caspase 8/metabolism , Cell Line , Cell Proliferation/drug effects , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Gene Expression Regulation , Indole Alkaloids/pharmacology , Kidney Tubules/cytology , Kidney Tubules/drug effects , Kidney Tubules/metabolism , MAP Kinase Kinase 7/genetics , MAP Kinase Kinase 7/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 10/metabolism , Plasmids/chemistry , Plasmids/metabolism , Rats , Trans-Activators/isolation & purification , Transfection , Viral Regulatory and Accessory Proteins , fas Receptor/genetics , fas Receptor/metabolism , Mitogen-Activated Protein Kinase Kinase Kinase 11
4.
J Immunol ; 193(12): 6103-13, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25378591

ABSTRACT

TLRs play an important role in mediating intestinal inflammation and homeostasis. Fas is best studied in terms of its function in apoptosis, but recent studies demonstrate that Fas signaling may mediate additional functions such as inflammation. The role of Fas, and the Fas ligand (FasL), in the intestine is poorly understood. The aim of this study was to evaluate potential cross-talk between TLRs and Fas/FasL system in intestinal epithelial cells (IECs). IECs were stimulated with TLR ligands, and expression of Fas and FasL was investigated. Treatment with TLR4 and TLR5 ligands, but not TLR2 and 9 ligands, increased expression of Fas and FasL in IECs in vitro. Consistent with this finding, expression of intestinal Fas and FasL was reduced in vivo in the epithelium of TLR4 knockout (KO), 5KO, and germ-free mice, but not in TLR2KO mice. Modulating Fas signaling using agonistic anti-Fas augmented TLR4- and TLR5-mediated TNF-α and IL-8 production by IECs. In addition, suppression of Fas in IECs reduced the ability of TLR4 and TLR5 ligands and the intestinal pathogens Salmonella typhimurium and Listeria monocytogenes to induce the expression of IL-8. In conclusion, this study demonstrates that extensive cross-talk in IECs occurs between the Fas and TLR signaling pathways, with the FasL/Fas system playing a role in TLR-mediated inflammatory responses in the intestine.


Subject(s)
Fas Ligand Protein/metabolism , Inflammation/metabolism , Intestinal Mucosa/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 5/metabolism , fas Receptor/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Cytokines/biosynthesis , Disease Models, Animal , Fas Ligand Protein/genetics , Gene Expression Regulation , HT29 Cells , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Intestinal Mucosa/immunology , Intestines/immunology , Intestines/microbiology , Ligands , Listeria monocytogenes , Mice , Mice, Knockout , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Salmonella typhimurium , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 5/agonists , Toll-Like Receptor 5/genetics , fas Receptor/agonists , fas Receptor/genetics
5.
Biosci Biotechnol Biochem ; 80(2): 250-6, 2016.
Article in English | MEDLINE | ID: mdl-26479732

ABSTRACT

Advanced glycation end-products (AGEs) are extremely accumulated in the retinal vascular and epithelial cells of diabetes mellitus (DM) patients, particularly with diabetic retinopathy (DR). To elucidate the pathogenesis of the AGE-induced toxicity to retinal epithelial cells, we investigated the role of Fas-Fas ligand (FasL) signaling and mitochondrial dysfunction in the AGE-induced apoptosis. Results demonstrated that the AGE-BSA- induced apoptosis of retinal ARPE-19 cells. And the AGE-BSA treatment caused mitochondrial dysfunction, via deregulating the B-cell lymphoma 2 (Bcl-2) signaling. Moreover, the Fas/FasL and its downstreamer Caspase 8 were promoted by the AGE-BSA treatment, and the exogenous α-Fas exacerbated the activation of Caspase 3/8. On the other side, the siRNA-mediated knockdown of Fas/FasL inhibited the AGE-BSA-induced apoptosis. Taken together, we confirmed the activation of Fas-FasL signaling and of mitochondrial dysfunction in the AGE-BSA-promoted apoptosis in retinal ARPE-19 cells, implying the important role of Fas-FasL signaling in the DR in DM.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/drug effects , Fas Ligand Protein/genetics , Glycation End Products, Advanced/pharmacology , Mitochondria/drug effects , fas Receptor/genetics , Caspase 3/genetics , Caspase 3/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cell Line , Epithelial Cells/metabolism , Fas Ligand Protein/agonists , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/metabolism , Gene Expression Regulation , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/pharmacology , Retina/drug effects , Retina/metabolism , Serum Albumin, Bovine/pharmacology , Signal Transduction , fas Receptor/agonists , fas Receptor/antagonists & inhibitors , fas Receptor/metabolism
6.
Cytokine ; 75(2): 228-33, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25481649

ABSTRACT

Rheumatoid Arthritis (RA) is a chronic inflammatory disease affecting synovial joints. Tumor necrosis factor (TNF) α is a key component of RA pathogenesis and blocking this cytokine is the most common strategy to treat the disease. Though TNFα blockers are very efficient, one third of the RA patients are unresponsive or present side effects. Therefore, the development of novel therapeutic approaches is required. RA pathogenesis is characterized by the hyperplasia of the synovium, closely associated to the pseudo-tumoral expansion of fibroblast-like synoviocytes (FLS), which invade and destroy the joint structure. Hence, depletion of RA FLS has been proposed as an alternative therapeutic strategy. The TNF family member Fas ligand (FasL) was reported to trigger apoptosis in FLS of arthritic joints by binding to its receptor Fas and therefore suggested as a promising candidate for targeting the hyperplastic synovial tissue. However, this cytokine is pleiotropic and recent data from the literature indicate that Fas activation might have a disease-promoting role in RA by promoting cell proliferation. Therefore, a FasL-based therapy for RA requires careful evaluation before being applied. In this review we aim to overview what is known about the apoptotic and non-apoptotic effects of Fas/FasL system and discuss its relevance in RA.


Subject(s)
Apoptosis/physiology , Arthritis, Rheumatoid/drug therapy , Fas Ligand Protein/metabolism , Synovial Membrane/pathology , fas Receptor/metabolism , Apoptosis/drug effects , Enzyme Activation/drug effects , Fas Ligand Protein/agonists , Fibroblasts/metabolism , Humans , Signal Transduction/immunology , Synovial Membrane/cytology , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/agonists
7.
Alcohol Clin Exp Res ; 38(3): 649-56, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24224890

ABSTRACT

BACKGROUND: Argininosuccinate synthase (ASS) is an enzyme shared by the urea cycle and the l-citrulline/nitric oxide (NO·) cycle. ASS is the rate-limiting enzyme in the urea cycle and along with nitric oxide synthase 2 (NOS2), it endows cells with the l-citrulline/NO· salvage pathway to continuously supply l-arginine from l-citrulline for sustained NO· generation. Thus, ASS conditions NO· synthesis by NOS2. Because of the relevance of NOS2 activation for liver injury, we examined the contribution of ASS to NO· generation and how it impacts liver injury. METHODS: Wild-type (WT) mice and Ass(+/-) mice (Ass(-/-) mice are lethal) were intraperitoneally injected with ethanol (EtOH) at a dose of 2.5 g/kg of body weight twice a day for 3 days. Two hours after the last dose of EtOH, mice were administered the agonistic Jo2 anti-mouse Fas monoclonal antibody (Ab) at a dose of 0.2 µg/g of body weight. Mice were sacrificed 8 hours after the Jo2 Ab injection. Markers of nitrosative and oxidative stress as well as liver damage were analyzed. RESULTS: EtOH plus Jo2 injection induced liver injury as shown by serum alanine aminotransferase and aspartate aminotransferase activity, liver pathology, TUNEL, and cleaved caspase-3 were lower in Ass(+/-) mice compared with WT mice, suggesting that ASS contributes to EtOH plus Jo2-mediated liver injury. CYP2E1 induction, glutathione depletion, and elevated thiobarbituric acid reactive substances were comparable in both groups of mice, suggesting that CYP2E1-mediated oxidative stress is not linked to ASS-induced liver injury. In contrast, NOS2 induction, 3-nitrotyrosine adducts formation and elevated nitrites, nitrates, and S-nitrosothiols were higher in livers from WT mice than from Ass(+/-) mice. CONCLUSION: Decreased nitrosative stress causes lower EtOH plus Jo2-induced liver injury in Ass(+/-) mice.


Subject(s)
Argininosuccinate Synthase/metabolism , Central Nervous System Depressants/adverse effects , Chemical and Drug Induced Liver Injury/enzymology , Ethanol/adverse effects , fas Receptor/agonists , Animals , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 CYP2E1/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress , Tumor Necrosis Factor-alpha/metabolism
8.
Adv Exp Med Biol ; 804: 203-17, 2014.
Article in English | MEDLINE | ID: mdl-24924176

ABSTRACT

The lungs are the most common site for the metastatic spread of osteosarcoma. Success in using chemotherapy to improve overall survival has reached a plateau. Understanding the biologic properties that permit osteosarcoma cells to grow in the lungs may allow the identification of novel therapeutic approaches-the goal being to alter the tumor cells' expression of cell surface proteins so that there is no longer compatibility with the metastatic niche. We have demonstrated that the Fas Ligand positive (FasL(+)) lung microenvironment eliminates Fas(+) osteosarcoma cells that metastasize to the lungs. Indeed, osteosarcoma lung metastases from patients are Fas(-), similar to what we found in several different mouse models. The Fas(+) cells are cleared from the lungs through apoptosis induced by the Fas signaling pathway following interaction of Fas on the tumor cell surface with the lung FasL. Blocking the Fas signaling pathway interferes with this process, allowing the Fas(+) cells to grow in the lungs. Our investigations show that Fas expression in osteosarcoma cells is regulated epigenetically by the micro-RNA miR-20a, encoded by the miR-17-92 cluster. Our studies support the feasibility of finding agents that can re-induce Fas expression as a novel therapeutic approach to treat osteosarcoma patients with lung metastases. We have identified two such agents, the histone deacetylase inhibitor entinostat and the chemotherapeutic agent gemcitabine (GCB). Aerosol GCB and oral entinostat induce the upregulation of Fas and the regression of established osteosarcoma lung metastases. Aerosol GCB was not effective in the FasL-deficient gld mouse confirming that the lung microenvironment was central to the success of this therapy. Our studies establish the critical role of the lung microenvironment in the metastatic process of osteosarcoma to the lungs and suggest an alternative focus for therapy, that is, incorporating the lung microenvironment as part of the treatment strategy against established osteosarcoma disease in the lungs.


Subject(s)
Bone Neoplasms/genetics , Fas Ligand Protein/genetics , Lung Neoplasms/genetics , Osteosarcoma/genetics , Tumor Microenvironment/drug effects , fas Receptor/genetics , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Epigenesis, Genetic , Fas Ligand Protein/agonists , Fas Ligand Protein/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Osteosarcoma/secondary , Pyridines/pharmacology , RNA, Long Noncoding , Signal Transduction , fas Receptor/agonists , fas Receptor/metabolism , Gemcitabine
9.
Tsitologiia ; 56(1): 77-83, 2014.
Article in Russian | MEDLINE | ID: mdl-25509146

ABSTRACT

The influence of UV-light (240-390 nm) in doses 151-3020 J/m2 on the nature of the death of lymphocytes cells of donor's blood (using markers of apoptotic and necrotic death of cells) and on the level of expression of the marker of apoptotic pre-preparation--CD95-receptor has been investigated. We have shown that UV-radiation increases expression of CD95-receptors which is caused mainly by de novo synthesis of the receptors. It has been revealed that during daily incubation of photo-modified lymphocytes (151 and 755 J/m2) without autological blood cell death occurs by receptor-involved apoptosis. Exposure to high doses of radiation (1510 and 3020 J/m2) causes massive necrotic death of immunocytes. The use of autologous blood plasma during incubation of photo-modified lymphocytes allows decreasing the number of both apoptotic and necrotic cells.


Subject(s)
Apoptosis/radiation effects , Lymphocytes/radiation effects , fas Receptor/biosynthesis , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers/metabolism , Cells, Cultured , Culture Media/chemistry , Culture Media/pharmacology , Dose-Response Relationship, Radiation , Gene Expression , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Lymphocytes/pathology , Necrosis/genetics , Necrosis/metabolism , Necrosis/pathology , Plasma , Ultraviolet Rays , fas Receptor/agonists , fas Receptor/genetics
10.
Cell Physiol Biochem ; 32(1): 111-20, 2013.
Article in English | MEDLINE | ID: mdl-23868143

ABSTRACT

BACKGROUND: Fas/CD95 is the best-studied member of the death receptor (DR) superfamily in the central nervous system where it can trigger cellular responses other than apoptosis, including the promotion of neurogenesis and neuritogenesis, stimulation of the progression of gliomas, and regulation of the immune response of astrocytes. METHODS: We have investigated the role of Fas/CD95 in the regulation of the proliferation of fetal astrocytes in vitro, as well as the signalling pathways involved. RESULTS: Fas/CD95 ligation stimulated the proliferation of primary fetal astrocytes, through a mechanism that depends on the activation of caspase 8 and subsequent phosphorylation of extracellular signal regulated kinase (ERK). Interestingly this proliferative effect is only observed with a low dose of the Fas/CD95 agonist. In contrast, when primary astrocytes are challenged with a high dose of the Fas/CD95 agonist significant cell death occurs. CONCLUSIONS: Our findings support that, besides its effects on cell survival, Fas/CD95 may play a complex and prominent role in the regulation of astrocyte proliferation during development.


Subject(s)
Astrocytes/cytology , Caspase 8/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , fas Receptor/metabolism , Animals , Astrocytes/metabolism , Cell Proliferation , Cells, Cultured , Female , Fetus/cytology , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction , fas Receptor/agonists
11.
Am J Physiol Lung Cell Mol Physiol ; 303(2): L141-51, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22610351

ABSTRACT

Excessive apoptosis and prolonged inflammation of alveolar cells are associated with the pathogenesis of pulmonary emphysema. We aimed to determine whether CD40 affects alveolar epithelial cells and endothelial cells, with regard to evoking apoptosis and inflammation. Mice were repeatedly treated with agonistic-anti CD40 antibody (Ab), with or without agonistic-anti Fas Ab, and evaluated for apoptosis and inflammation in lungs. Human pulmonary microvascular endothelial cells and alveolar epithelial cells were treated with agonistic anti-CD40 Ab and/or anti-Fas Ab to see their direct effect on apoptosis and secretion of proinflammatory molecules in vitro. Furthermore, plasma soluble CD40 ligand (sCD40L) level was evaluated in patients with chronic obstructive pulmonary disease (COPD). In mice, inhaling agonistic anti-CD40 Ab induced moderate alveolar enlargement. CD40 stimulation, in combination with anti-Fas Ab, induced significant emphysematous changes and increased alveolar cell apoptosis. CD40 stimulation also enhanced IFN-γ-mediated emphysematous changes, not via apoptosis induction, but via inflammation with lymphocyte accumulation. In vitro, Fas-mediated apoptosis was enhanced by CD40 stimulation and IFN-γ in endothelial cells and by CD40 stimulation in epithelial cells. CD40 stimulation induced secretion of CCR5 ligands in endothelial cells, enhanced with IFN-γ. Plasma sCD40L levels were significantly increased in patients with COPD, inversely correlating to the percentage of forced expiratory volume in 1 s and positively correlating to low attenuation area score by CT scan, regardless of smoking history. Collectively CD40 plays a contributing role in the development of pulmonary emphysema by sensitizing Fas-mediated apoptosis in alveolar cells and increasing the secretion of proinflammatory chemokines.


Subject(s)
Apoptosis , CD40 Antigens/metabolism , Pulmonary Emphysema/metabolism , fas Receptor/metabolism , Aged , Animals , Antibodies/pharmacology , CD40 Antigens/agonists , CD40 Antigens/immunology , CD40 Ligand/blood , CD40 Ligand/metabolism , Caspase 3/metabolism , Cells, Cultured , Chemokines, CC/metabolism , Endothelial Cells/metabolism , Female , Forced Expiratory Volume , Humans , Interferon-gamma/metabolism , Lung/blood supply , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Microvessels/pathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Emphysema/pathology , Receptors, CCR5/metabolism , fas Receptor/agonists , fas Receptor/immunology
12.
J Hepatol ; 55(6): 1272-80, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21703207

ABSTRACT

BACKGROUND & AIMS: Apoptosis is crucially involved in acute and chronic liver injury, including viral, cholestatic, toxic, and metabolic liver disease. Additionally, dysregulation of apoptosis signaling pathways has been implicated in hepatocarcinogenesis. The most prominent members of the apoptosis-mediating tumor necrosis factor receptor superfamily are the TNF-R1 (CD120a) and the CD95 (Apo-1/Fas) receptor. Although extensively studied, the intracellular signaling events in hepatocytes are only incompletely understood. METHODS: To examine the role of the caspase-8 homolog cellular FLICE-inhibitory protein (c-FLIP) in liver injury, we generated mice with hepatocyte specific deletion of c-FLIP. Three models of acute liver injury were employed: the agonistic anti-CD95 antibody Jo2, d-galactosamine and LPS (GalN/LPS), and concanavalin A. RESULTS: Conditional ablation of c-FLIP in hepatocytes augmented liver injury and cell death in all three models of liver injury. CD95- and GalN/LPS-induced liver injury was ameliorated by a pancaspase inhibitor, while ConA-induced injury was unaffected by caspase inhibition. Augmented activation of the MAPK JNK was observed in parallel to liver injury in c-FLIP knockout mice in all injury models; however, inhibition of JNK only affected TNF- and ConA-mediated injury. CONCLUSIONS: In summary, c-FLIP is a central regulator of cell death in hepatocytes, involving increased activation of caspases and the MAPK JNK.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Receptors, Death Domain/metabolism , Animals , Anthracenes/pharmacology , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspases/metabolism , Concanavalin A/toxicity , Female , Galactosamine/toxicity , Hepatocytes/drug effects , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Mice , Mice, Knockout , fas Receptor/agonists
13.
Eur J Immunol ; 40(11): 3043-53, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21061436

ABSTRACT

The size of the peripheral T-lymphocyte compartment is governed by complex homeostatic mechanisms that balance T-cell proliferation and death. Proliferation and survival signals are mediated in part by recurrent self-peptide/MHC-TCR interactions and signaling by the common γ chain-containing cytokine receptors, including those for IL-7 and IL-15. We have previously shown that the death receptor Fas (CD95/APO-1) regulates apoptosis in response to repeated TCR stimulation, whereas the Bcl-2 homology domain 3-only protein Bim mediates cytokine withdrawal-induced apoptosis. We therefore reasoned that these two molecules might cooperate in the regulation of homeostatic proliferation. In this study, we observe that the combined loss of Fas and Bim synergistically enhances the accumulation of T cells in lymphopenic host mice, and this is particularly pronounced for the unusual CD4(-) CD8(-) TCRαß(+) T cells that are characteristic of Fas-deficient (Fas(lpr/lpr) ) mice. Our findings demonstrate that these CD4(-) CD8(-) TCRαß(+) T cells arise from homeostatic proliferation of CD8(+) T cells. These studies also underscore the profound rate of baseline T-cell proliferation that likely occurs in wild-type mice even in the absence of foreign antigen, and the consequent need for its coordinated regulation by multiple death-signaling pathways.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Homeostasis/immunology , Membrane Proteins/immunology , Proto-Oncogene Proteins/immunology , fas Receptor/immunology , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/agonists , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Histocompatibility Antigens/immunology , Homeostasis/genetics , Interleukin-15/genetics , Interleukin-15/immunology , Interleukin-7/genetics , Interleukin-7/immunology , Membrane Proteins/agonists , Membrane Proteins/genetics , Mice , Mice, Knockout , Peptides/immunology , Proto-Oncogene Proteins/agonists , Proto-Oncogene Proteins/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction/genetics , Signal Transduction/immunology , fas Receptor/agonists , fas Receptor/genetics
14.
J Immunol ; 183(5): 3278-84, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19641134

ABSTRACT

The serine-threonine kinase RIP1 was originally identified through its ability to bind to the death domain of Fas (CD95). RIP1 has been shown to be recruited to the Fas death-inducing signaling complex (DISC) and is required for the induction of necrotic cell death. In this study, we show that in Jurkat T lymphocytes, RIP1 is also necessary for the most efficient activation of downstream caspases by Fas when treated with membrane-bound Fas ligand, but not with agonistic Abs or cross-linked soluble Fas ligand. RIP1 participates in the Fas-associated death domain protein-mediated recruitment of caspase-8 to the Fas receptor complex in a manner that promotes caspase-8 activation. Cross-linking Abs, such as CH11, bypass the requirement for RIP1 in caspase activation by initiating larger, though less efficient, DISC complexes, while membrane-bound Fas ligand initiates a smaller but more efficient DISC that functions, in part, by effectively incorporating more RIP1 into the complex. Consequently, RIP1 is likely a more integral part of physiological signaling through the Fas/CD95 receptor complex than previously recognized; at least when the signal is mediated by full-length membrane-bound FasL. Cross-linked soluble FasL, which also occurs physiologically, behaves similarly to the CH11 Ab, and may therefore be more likely to initiate nonapoptotic Fas signaling due to less RIP1 in the receptor complex. Thus, agonists that bind the same Fas receptor initiate mechanistically distinct pathways resulting in differential cytotoxicity.


Subject(s)
Caspase 8/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Fas Ligand Protein/metabolism , Membrane Proteins/metabolism , Nuclear Pore Complex Proteins/physiology , RNA-Binding Proteins/physiology , Antibodies/metabolism , Cell Death/immunology , Cross-Linking Reagents/metabolism , Cytotoxicity, Immunologic , Enzyme Activation/immunology , Fas Ligand Protein/immunology , Fas Ligand Protein/toxicity , Humans , Immunity, Innate , Immunoglobulin M/metabolism , Jurkat Cells , Membrane Proteins/immunology , Membrane Proteins/toxicity , Nuclear Pore Complex Proteins/deficiency , Nuclear Pore Complex Proteins/metabolism , Oligopeptides , Peptides/immunology , RNA-Binding Proteins/metabolism , fas Receptor/agonists , fas Receptor/immunology , fas Receptor/metabolism
15.
Biochemistry ; 49(29): 6263-75, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20565132

ABSTRACT

In recent years, 4-hydroxy-2-nonenal (4-HNE) has emerged as an important second messenger in cell cycle signaling. Here, we demonstrate that 4-HNE induces signaling for apoptosis via both the Fas-mediated extrinsic and the p53-mediated intrinsic pathways in HepG2 cells. 4-HNE induces a Fas-mediated DISC independent apoptosis pathway by activating ASK1, JNK, and caspase-3. Parallel treatment of 4-HNE to HepG2 cells also induces apoptosis by the p53 pathway through activation of Bax, p21, JNK, and caspase-3. Exposure of HepG2 cells to 4-HNE leads to the activation of both Fas and Daxx, promotes the export of Daxx from the nucleus to cytoplasm, and facilitates Fas-Daxx binding. Depletion of Daxx by siRNA results in the potentiation of apoptosis, indicating that Fas-Daxx binding in fact is inhibitory to Fas-mediated apoptosis in cells. 4-HNE-induced translocation of Daxx is also accompanied by the activation and nuclear accumulation of HSF1 and up-regulation of heat shock protein Hsp70. All these effects of 4-HNE in cells can be attenuated by ectopic expression of hGSTA4-4, the isozyme of glutathione S-transferase with high activity for 4-HNE. Through immunoprecipitation and liquid chromatography-tandem mass spectrometry, we have demonstrated the covalent binding of 4-HNE to Daxx. We also demonstrate that 4-HNE modification induces phosphorylation of Daxx at Ser668 and Ser671 to facilitate its cytoplasmic export. These results indicate that while 4-HNE exhibits toxicity through several mechanisms, in parallel it evokes signaling for defense mechanisms to self-regulate its toxicity and can simultaneously affect multiple signaling pathways through its interactions with membrane receptors and transcription factors/repressors.


Subject(s)
Aldehydes/pharmacology , Apoptosis/drug effects , Death Domain Receptor Signaling Adaptor Proteins/metabolism , fas Receptor/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Co-Repressor Proteins , Enzyme Activation , Humans , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Molecular Chaperones , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Signal Transduction/drug effects , fas Receptor/agonists
16.
Biochem Biophys Res Commun ; 391(1): 575-81, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19941832

ABSTRACT

C(2)-ceramide (C(2)-cer) and binding of the CD95/APO-1/FAS (aCD95) receptor are acknowledged inducers of apoptosis. In spite of that, their effects on the endoplasmic reticulum (ER) and mitochondria during early phases of apoptotic onset are poorly characterized. Here, by employing various approaches, we followed structural and functional modifications of these organelles at the beginning of cellular demise. In detail, we observed that C(2)-cer, but not CD95 activation, markedly modifies the morphology of the ER and promotes Ca(2+) release. Accordingly, mitochondria of C(2)-cer-treated, but not of CD95-stimulated, cells are fragmented, show reduced Ca(2+) uptake, and collapsed membrane potential (DeltaPsi(m)). Most notably, C(2)-cer-mediated morphological aberrations of the ER are prevented neither by the pan-caspase inhibitor Z-VADfmk nor by the cell cytoskeleton dissembler cytochalasin-D, while on the contrary they are reduced by incubation in the presence of the intracellular Ca(2+) chelator 1,2-bis(O-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA). We conclude that initiation of apoptosis via the intrinsic (i.e. C(2)-cer mediated) pathway causes an early structural and functional alteration of both ER and mitochondria, thus underlying a final "non return" point in the apoptotic pathway.


Subject(s)
Apoptosis , Endoplasmic Reticulum/ultrastructure , Mitochondria/ultrastructure , Sphingosine/analogs & derivatives , fas Receptor/agonists , Amino Acid Chloromethyl Ketones/pharmacology , Calcium/metabolism , Caspase Inhibitors , Chelating Agents/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Sphingosine/metabolism , fas Receptor/metabolism
17.
Biochem Biophys Res Commun ; 399(2): 198-202, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20643097

ABSTRACT

Activation of T-cells triggers store-operated Ca(2+) entry, which begins a signaling cascade leading to induction of appropriate gene expression and eventually lymphocyte proliferation and differentiation. The simultaneous enhancement of Fas ligand gene expression in activated cells allows the immune response to be limited by committing the activated cells to apoptosis. In apoptotic cells the store-operated calcium entry is significantly inhibited. It has been documented that moderate activation of Fas receptor may cause reversible inhibition of store-operated channels by ceramide released from hydrolyzed sphingomyelin. Here we show that activation of Fas receptor in T-cells results in caspase-dependent decrease of cellular STIM1 and Orai1 protein content. This effect may be responsible for the substantial inhibition of Ca(2+) entry into Jurkat cells undergoing apoptosis. In turn, this inhibition might prevent overloading of cells with calcium and protect them against necrosis.


Subject(s)
Apoptosis , Calcium Signaling , Calcium/metabolism , Caspases/metabolism , T-Lymphocytes/immunology , Antibodies/immunology , Calcium Channels/metabolism , Caspase Inhibitors , Humans , Jurkat Cells , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , ORAI1 Protein , Stromal Interaction Molecule 1 , T-Lymphocytes/drug effects , fas Receptor/agonists , fas Receptor/metabolism
18.
Bol Med Hosp Infant Mex ; 77(4): 186-194, 2020.
Article in English | MEDLINE | ID: mdl-32713953

ABSTRACT

Background: Acute lymphoblastic leukemia (ALL) is an aggressive malignant disease with high prevalence in pediatric patients. It has been shown that the downregulation of Fas expression is correlated with an inadequate response in ALL, although these mechanisms are still not well understood. Several reports demonstrated that hypoxia is involved in dysfunctional apoptosis. Yin-Yang-1 (YY1) transcription factor is involved in resistance to apoptosis, tumor progression, and it is increased in different types of cancer, including leukemia. The regulatory mechanism underlying YY1 expression in leukemia is still not understood, but it is known that YY1 negatively regulates Fas expression. The study aimed to evaluate the effect of YY1 on Fas expression under hypoxic conditions in ALL. Methods: Leukemia cell line RS4; 11 was cultured under normoxic and hypoxic conditions. YY1, Fas receptor, and hypoxia-inducible factor (HIF-1α) expression were analyzed. After treatment with a Fas agonist (DX2), apoptosis was analyzed through the detection of active caspase 3. Data were analyzed using Pearson's correlation. Results: Leukemia cells co-expressed both HIF-1α and YY1 under hypoxia, which correlated with a downregulation of Fas expression. During hypoxia, the levels of apoptosis diminished after DX2 treatment. The analysis revealed that patients with high levels of HIF-1α also express high levels of YY1 and low levels of Fas. Conclusions: These results suggest that YY1 negatively regulates the expression of the Fas receptor, which could be involved in the escape of leukemic cells from the immune response contributing to the ALL pathogenesis.


Introducción: La leucemia linfoblástica aguda (LLA) es una enfermedad con alta prevalencia en la población pediátrica. El mecanismo por el cual el receptor de Fas participa en la regulación inmunitaria en los tumores es desconocido, pero se sabe que está subexpresado en LLA. El factor de transcripción Ying-Yang-1 (YY1) está involucrado en la resistencia a la apoptosis y la progresión tumoral; se encuentra aumentado en diferentes tumores, incluida la LLA. Aunque los mecanismos que regulan la expresión de YY1 en LLA son desconocidos, se sabe que YY1 regula la expresión del receptor de Fas. El objetivo de este trabajo fue evaluar el efecto de YY1 en la expresión de Fas en condiciones de hipoxia en la LLA. Métodos: Se cultivaron células RS4;11 en condiciones de hipoxia y se analizó la expresión de YY1, receptor de Fas y HIF-1α. La apoptosis fue inducida usando un agonista de Fas (DX2) y se analizó con la detección de caspasa 3 activa. Los datos se analizaron mediante correlación de Pearson. Resultados: Las células RS4;11 coexpresaron HIF-1αy YY1 en hipoxia, lo cual correlaciona con una baja expresión de Fas. La apoptosis se encontró disminuida durante condiciones de hipoxia, después del tratamiento con DX2. El análisis bioinformático mostró que los pacientes con altos niveles de HIF-1αpresentan YY1 elevado y bajos niveles del receptor de Fas. Conclusiones: Estos resultados sugieren que YY1 regula negativamente la expresión del receptor de Fas, lo cual podría estar involucrado en el escape de las células leucémicas a la respuesta inmunitaria, contribuyendo a la patogénesis de la LLA.


Subject(s)
Apoptosis/physiology , Cell Hypoxia/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , YY1 Transcription Factor/metabolism , fas Receptor/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Child , Down-Regulation , Gene Expression , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immune Evasion , Immunologic Surveillance , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Hypoxia/physiology , YY1 Transcription Factor/genetics , fas Receptor/agonists
19.
Immunology ; 128(1 Suppl): e589-99, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19740320

ABSTRACT

Within the lesions of inflammatory myopathies, muscle fibres and invading mononuclear cells express Fas and Fas ligand (FasL), respectively. However, the roles of the Fas/FasL interaction in the pathogenesis of inflammatory myopathies are not fully understood. In the present study, we investigated the roles of proinflammatory cytokines and the Fas/FasL system in the pathogenesis of inflammatory myopathies. In vitro culturing of muscle cells with the proinflammatory cytokines interferon-gamma, tumour necrosis factor-alpha, and interleukin (IL)-1beta synergistically increased Fas expression, susceptibility to Fas-mediated apoptosis, and the expression of cytoplasmic caspases 8 and 3. In addition, culturing of muscle cells with activated CD4(+) T cells induced muscle cell apoptosis, which was partially inhibited by anti-FasL antibody. We also tested the possibility that T helper (Th) 17, which is an IL-17-producing helper T-cell subset that plays crucial roles in autoimmune and inflammatory responses, participates in the pathogenesis of inflammatory myopathies. Interestingly, in vitro culturing of dendritic cells with anti-Fas immunoglobulin M (IgM) or activated CD4(+) T cells induced the expression of mRNA for IL-23p19, but not for IL-12p35, in addition to proinflammatory cytokines. Furthermore, IL-23p19 and IL-17 mRNAs were detected in the majority of biopsy samples from patients with inflammatory myopathies. Taken together, these results suggest that proinflammatory cytokines enhance Fas-mediated apoptosis of muscle cells, and that the Fas/FasL interaction between invading dendritic cells and CD4(+) T cells induces local production of IL-23 and proinflammatory cytokines, which can promote the proliferation of Th17 cells and enhance Fas-mediated apoptosis of muscle cells, respectively.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fas Ligand Protein/immunology , Muscle Cells/immunology , Myositis/immunology , fas Receptor/metabolism , Apoptosis/drug effects , Apoptosis/immunology , CD4-Positive T-Lymphocytes/drug effects , Caspase 3/drug effects , Caspase 3/immunology , Caspase 8/drug effects , Caspase 8/metabolism , Cell Line , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Fas Ligand Protein/metabolism , Humans , Interferon-gamma/pharmacology , Interleukin-12 Subunit p35/metabolism , Interleukin-17/metabolism , Interleukin-1beta/pharmacology , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukin-23 Subunit p19/metabolism , Muscle Cells/drug effects , Myositis/pathology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/agonists
20.
Int J Oncol ; 34(3): 717-27, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19212677

ABSTRACT

Glioblastoma is the most common primary brain tumor, characterized by its resistance to treatments. To define efficient therapy, the origin of tumor-forming cells needs to be elucidated in order to search for new therapeutic pathways. The objective of this study was to determine the different cell populations constituting a human glioblastoma cell line, U-87 MG and their sensitivity to apoptosis induced through the activation of Fas, a membranous death receptor. By a cell sorting method, the sedimentation field flow fractionation, two major cell subpopulations were identified, a most differentiated cell fraction, containing large and adherent cells, sensitive to Fas-induced apoptosis and another one, characterized by small cells forming aggregates, expressing CD133, a marker of stem cells and more resistant to Fas-activated apoptosis. By using a selective method of culture, adapted for neural stem cell cultures, we have verified that the U-87 MG cell line contained cancer stem cells similar to the immature ones obtained by the cell sorting method. Interestingly, while these tumor stem cells, expressing CD133, were resistant to Fas-induced apoptosis, monomeric form of Fas protein was detected predominantly in these cells. In contrast, the most mature cells, responsive to Fas-activated apoptosis, collected in another cell fraction, contained oligomeric aggregates of Fas protein, a pre-signalling form of the Fas receptor, essential for the initiation of apoptosis through its activation. These results suggest that these immature stem cells in glioma could be an important factor of resistance to chemotherapy requiring apoptosis through Fas signalling system. Indeed, future strategies of treatment, inducing differentiation of these stem cells need to be considered to enhance therapeutic efficiency.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis/physiology , Brain Neoplasms/pathology , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , fas Receptor/agonists , AC133 Antigen , Antibodies, Monoclonal/immunology , Antigens, CD/biosynthesis , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Flow Cytometry , Glioblastoma/drug therapy , Glycoproteins/biosynthesis , Humans , Neoplastic Stem Cells/metabolism , Peptides , fas Receptor/biosynthesis , fas Receptor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL