Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
Article in English | MEDLINE | ID: mdl-29870789

ABSTRACT

Based on the assumed oestrogenic and apoptotic properties of soya isoflavones (genistein, daidzein), and following the current OECD test-guidelines and principle of 3Rs, we have studied the potential toxicity of phytochemicals on the zebrafish embryos test (ZFET). For this purpose, zebrafish embryos at 2-3 h post-fertilisation (hpf) were exposed to both soya isoflavones (from 1.25 mg/L to 20 mg/L) and assayed until 96 hpf. Lethal and sub-lethal endpoints (mortality, hatching rates and malformations) were estimated in the ZFET, which was expanded to potential gene expression markers, determining the lowest observed effect (and transcriptional) concentrations (LOEC, LOTEC), and the no-observable effect (and transcriptional) concentrations (NOEC, NOTEC). The results revealed that genistein is more toxic (LC50-96 hpf: 4.41 mg/L) than daidzein (over 65.15 mg/L). Both isoflavones up-regulated the oestrogen (esrrb) and death receptors (fas) and cyp1a transcript levels. Most thyroid transcript signals were up-regulated by genistein (except for thyroid peroxidase/tpo), and the hatching enzyme (he1a1) was exclusively up-regulated by daidzein (from 1.25 mg/L onwards). The ZFET proved suitable for assessing toxicant effects of both isoflavones and potential disruptions (i.e. oestrogenic, apoptotic, thyroid, enzymatic) during the embryogenesis and the endotrophic larval period.


Subject(s)
Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Genistein/adverse effects , Isoflavones/adverse effects , Phytoestrogens/adverse effects , Thyroid Gland/metabolism , Animals , Apoptosis , Cytochrome P-450 CYP1A1/chemistry , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Dietary Supplements/adverse effects , Ectogenesis , Embryo, Nonmammalian/enzymology , Endocrine Disruptors/adverse effects , Endocrine Disruptors/metabolism , Genistein/metabolism , Isoflavones/metabolism , Larva/enzymology , Larva/growth & development , Larva/metabolism , Lethal Dose 50 , Receptors, Estrogen/chemistry , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Seeds/chemistry , Signal Transduction , Glycine max/chemistry , Thyroid Gland/embryology , Thyroid Gland/enzymology , Toxicity Tests, Acute , Zebrafish , fas Receptor/agonists , fas Receptor/chemistry , fas Receptor/metabolism
4.
Toxicol Sci ; 162(2): 396-405, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29228388

ABSTRACT

Tissue factor (TF) is the primary activator of the blood coagulation cascade. Liver parenchymal cells (ie, hepatocytes) express TF in a molecular state that lacks procoagulant activity. Hepatocyte apoptosis is an important feature of acute and chronic liver diseases, and Fas-induced apoptosis increases hepatocyte TF procoagulant activity in vitro. We determined the impact of a pan-caspase inhibitor, IDN-7314, on hepatocyte TF activity in vitro and TF-mediated coagulation in vivo. Treatment of primary mouse hepatocytes with the Fas death receptor ligand (Jo2, 0.5 µg/ml) for 8 h increased hepatocyte TF procoagulant activity and caused release of TF-positive microvesicles. Pretreatment with 100 nM IDN-7314 abolished Jo2-induced caspase-3/7 activity and significantly reduced hepatocyte TF procoagulant activity and release of TF-positive microvesicles. Treatment of wild-type C57BL/6 mice with a sublethal dose of Jo2 (0.35 mg/kg) for 4.5 h increased coagulation, measured by a significant increase in plasma thrombin-antithrombin and TF-positive microvesicles. Total plasma microvesicle-associated TF activity was reduced in mice lacking hepatocyte TF; suggesting TF-positive microvesicles are released from the apoptotic liver. Fibrin(ogen) deposition increased in livers of Jo2-treated wild-type mice and colocalized primarily with cleaved caspase-3-positive hepatocytes. Pretreatment with IDN-7314 reduced caspase-3 activation, prevented the procoagulant changes in Jo2-treated mice, and reduced hepatocellular injury. Overall, the results indicate a central role for caspase activity in TF-mediated activation of coagulation following apoptotic liver injury. Moreover, the results suggest that liver-selective caspase inhibition may be a putative strategy to limit procoagulant and prothrombotic changes in patients with chronic liver disease.


Subject(s)
Apoptosis/drug effects , Blood Coagulation/drug effects , Caspase Inhibitors/pharmacology , Hepatocytes/drug effects , Liver/blood supply , Thromboplastin/metabolism , Animals , Caspase 3/metabolism , Hepatocytes/pathology , Liver/metabolism , Male , Mice, Inbred C57BL , Microvessels/drug effects , Microvessels/pathology , Primary Cell Culture , fas Receptor/agonists
5.
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
6.
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
7.
Toxicol Lett ; 258: 71-86, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27313093

ABSTRACT

The role of reactive metabolites and inflammatory stress has been largely evoked in idiosyncratic hepatotoxicity of diclofenac (DCF); however mechanisms remain poorly understood. We aimed to evaluate the influence of liver cell phenotype on the hepatotoxicity of DCF combined or not with TNF-α using differentiated and undifferentiated HepaRG cells, and for comparison, HepG2 cells. Our results demonstrate that after a 24h-treatment metabolizing HepaRG cells were less sensitive to DCF than their undifferentiated non-metabolizing counterparts as shown by lower oxidative and endoplasmic reticulum stress responses and lower activation of caspase 9. Differentiated HepaRG cells were also less sensitive than HepG2 cells. Their lower sensitivity to DCF was related to their high content in glutathione transferases. DCF-induced apoptotic effects were potentiated by TNF-α only in death receptor-expressing differentiated HepaRG and HepG2 cells and were associated with marked activation of caspase 8. TNF-α co-treatment did not aggravate DCF-induced cholestatic features. Altogether, our results demonstrate that (i) lower sensitivity to DCF of differentiated HepaRG cells compared to their non-metabolically active counterparts was related to their high detoxifying capacity, giving support to the higher sensitivity of nonhepatic tissues than liver to this drug; (ii) TNF-α-potentiation of DCF cytotoxicity occurred only in death receptor-expressing cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Diclofenac/pharmacology , Drug Resistance , Hepatocytes/drug effects , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/agonists , Anti-Inflammatory Agents, Non-Steroidal/agonists , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Biotransformation/drug effects , Cell Differentiation , Cell Line , Cells, Cultured , Diclofenac/agonists , Diclofenac/metabolism , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/metabolism , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Inhibitory Concentration 50 , Kinetics , Microscopy, Phase-Contrast , Oxidative Stress/drug effects , fas Receptor/metabolism
8.
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
9.
FEBS Lett ; 589(23): 3527-33, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26484594

ABSTRACT

Extrinsic apoptosis is initiated by recognition and clustering of the single-pass transmembrane proteins Fas ligand and Fas expressed at the surface of closely apposed lymphocytes and target cells, respectively. Since Fas-mediated death response was mainly studied with soluble antibodies, the mobility constraints for receptor activation by a membrane embedded agonist is not well understood. We explored this influence by stimulating apoptosis on functionalized supported lipid bilayers, where we quantified agonist mobility by z-scan fluorescence correlation spectroscopy. Using different lipid compositions, we show that the apoptotic response correlates with increased lateral mobility of the agonist in the lipid bilayer.


Subject(s)
Apoptosis , Lipid Bilayers/metabolism , Movement , fas Receptor/agonists , fas Receptor/metabolism , HEK293 Cells , Humans , Membrane Fluidity
10.
PLoS One ; 10(5): e0125727, 2015.
Article in English | MEDLINE | ID: mdl-25933104

ABSTRACT

Arctigenin (ARG) has been previously reported to exert high biological activities including anti-inflammatory, antiviral and anticancer. In this study, the anti-tumor mechanism of ARG towards human hepatocellular carcinoma (HCC) was firstly investigated. We demonstrated that ARG could induce apoptosis in Hep G2 and SMMC7721 cells but not in normal hepatic cells, and its apoptotic effect on Hep G2 was stronger than that on SMMC7721. Furthermore, the following study showed that ARG treatment led to a loss in the mitochondrial out membrane potential, up-regulation of Bax, down-regulation of Bcl-2, a release of cytochrome c, caspase-9 and caspase-3 activation and a cleavage of poly (ADP-ribose) polymerase in both Hep G2 and SMMC7721 cells, suggesting ARG-induced apoptosis was associated with the mitochondria mediated pathway. Moreover, the activation of caspase-8 and the increased expression levels of Fas/FasL and TNF-α revealed that the Fas/FasL-related pathway was also involved in this process. Additionally, ARG induced apoptosis was accompanied by a deactivation of PI3K/p-Akt pathway, an accumulation of p53 protein and an inhibition of NF-κB nuclear translocation especially in Hep G2 cells, which might be the reason that Hep G2 was more sensitive than SMMC7721 cells to ARG treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Furans/pharmacology , Gene Expression Regulation, Neoplastic , Lignans/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Apoptosis , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Fas Ligand Protein/agonists , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Hep G2 Cells , Humans , Mitochondria/metabolism , Mitochondria/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Organ Specificity , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , fas Receptor/agonists , fas Receptor/genetics , fas Receptor/metabolism
11.
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
12.
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
13.
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
14.
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
15.
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
16.
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
17.
Food Chem Toxicol ; 59: 281-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23770345

ABSTRACT

Although numerous studies have shown the cancer-preventive properties of butylidenephthalide (BP), there is little report of BP affecting human prostate cancer cells. In the present study, proteomic-based approaches were used to elucidate the anticancer mechanism of BP in LNCaP human prostate cancer cells. BP treatment decreased the viability of LNCaP human prostate cancer cells in a concentration- and time-dependent manner, which was correlated with G0/G1 phase cell cycle arrest. Increased cell cycle arrest was associated with a decrease in the level of CCND1, CDK2, and PCNA proteins and an increase in the level of CDKN2A, CDKN1A, and SFN proteins. Proteomic studies revealed that among 48 differentially expressed proteins, 25 proteins were down-regulated and 23 proteins were up-regulated and these proteins fall into one large protein protein interaction network. Among these proteins, FAS, AIFM1, BIK, CYCS, SFN, PPP2R1A, CALR, HSPA5, DDIT3, and ERN1 are apoptosis and endoplasmic reticulum (ER) stress associated proteins. Proteomic data suggested that multiple signaling pathways including FAS-dependent pathway, mitochondrial pathway, and ER stress pathway are involved in the apoptosis induced by BP.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Models, Biological , Phthalic Anhydrides/pharmacology , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Adenocarcinoma/enzymology , Adenocarcinoma/metabolism , Apoptosis Regulatory Proteins/agonists , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/agonists , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Electrophoresis, Gel, Two-Dimensional , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Humans , Male , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/metabolism , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Peptide Mapping , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Resting Phase, Cell Cycle/drug effects , fas Receptor/agonists , fas Receptor/chemistry , fas Receptor/metabolism
18.
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
20.
Cell Death Differ ; 19(7): 1187-95, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22261618

ABSTRACT

Receptor agonism remains poorly understood at the molecular and mechanistic level. In this study, we identified a fully human anti-Fas antibody that could efficiently trigger apoptosis and therefore function as a potent agonist. Protein engineering and crystallography were used to mechanistically understand the agonistic activity of the antibody. The crystal structure of the complex was determined at 1.9 Å resolution and provided insights into epitope recognition and comparisons with the natural ligand FasL (Fas ligand). When we affinity-matured the agonist antibody, we observed that, surprisingly, the higher-affinity antibodies demonstrated a significant reduction, rather than an increase, in agonist activity at the Fas receptor. We propose and experimentally demonstrate a model to explain this non-intuitive impact of affinity on agonist antibody signalling and explore the implications for the discovery of therapeutic agonists in general.


Subject(s)
Antibodies/immunology , fas Receptor/agonists , Antibodies/genetics , Apoptosis/drug effects , Binding Sites , Crystallography, X-Ray , Fas Ligand Protein/pharmacology , HeLa Cells , Humans , Jurkat Cells , Kinetics , Mutagenesis , Protein Engineering , Protein Structure, Tertiary , Signal Transduction , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , fas Receptor/immunology , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...