Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Neurotox Res ; 39(4): 1103-1115, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33689146

ABSTRACT

Although many studies have reported toxic effects of cadmium (Cd) and lead (Pb) in the central nervous system, few studies have investigated the combined toxicity of Cd and Pb. The mechanisms by which these combined heavy metals induce toxicity, as well as effective means to exert neuroprotection from these agents, remain poorly understood. To investigate the protective effects of alpha-lipoic acid (α-LA) on Cd- and/or Pb-induced cortical damage in rats, 48 Sprague-Dawley rats were exposed to drinking water containing 50 mg/L of Cd and/or 300 mg/L of Pb for 12 weeks, in the presence or absence of α-LA co-treatment (50 mg/kg) via gavage. We observed that exposure to Cd and/or Pb decreased the brain weight/body weight ratio and increased Cd and/or Pb contents as well as ultrastructural damage to the cerebral cortex. Cd and/or Pb also induced endoplasmic-reticulum (ER) stress and activated Fas (CD95/APO-1)/Fas ligand (FasL) and mitochondrial apoptotic pathways. Furthermore, co-treatment of Cd and Pb further exacerbated part of these phenotypes than treatment of Cd or Pb alone. However, simultaneous supplementation with α-LA attenuated Cd and/or Pb-induced neurotoxicity by increasing the brain weight/body weight ratio, reducing Cd and/or Pb contents, ameliorating both nuclear/mitochondrial damage and ER stress, and attenuating activation of Fas/FasL and mitochondrial apoptotic pathways. Collectively, our results indicate that the accumulation of Cd and/or Pb causes cortical damage and that α-LA exerts protection against Cd- and/or Pb-induced neurotoxicity. These findings highlight that α-LA may be exploited for the treatment and prevention of Cd- and/or Pb-induced neurotoxicity.


Subject(s)
Cadmium/toxicity , Cerebral Cortex/drug effects , Endoplasmic Reticulum Stress/drug effects , Fas Ligand Protein/antagonists & inhibitors , Lead/toxicity , Thioctic Acid/pharmacology , fas Receptor/antagonists & inhibitors , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Endoplasmic Reticulum Stress/physiology , Fas Ligand Protein/metabolism , Female , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Rats , Rats, Sprague-Dawley , fas Receptor/metabolism
2.
Stem Cell Res Ther ; 10(1): 368, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31796122

ABSTRACT

BACKGROUND: Gingiva-derived mesenchymal stem cells (GMSCs) obtained multipotent differentiation and immunomodulatory properties. However, collecting healthy gingival tissues may be challenging in the clinical situation. Thus, in our present study, we aim to evaluate whether the immunomodulatory capacity of gingiva-derived mesenchymal stem cells from inflamed gingival tissues (iGMSCs) is impaired and find a way to rescue their deficient properties. METHODS: We compared the immunomodulation capacity of GMSCs and iGMSCs using an in vitro co-culture system and a mouse colitis model. T cell apoptosis, T helper 17 (Th17), and regulatory T (Treg) cell differentiation were detected by flow cytometry analysis. RESULTS: We demonstrated that iGMSCs obtained a decreased immunomodulatory capacity compared with GMSCs. Acetylsalicylic acid (ASA) pretreatment was able to rescue iGMSCs' impaired immunomodulatory properties. Mechanistically, ASA was capable of upregulating the expression of Fas ligand (FasL) in iGMSCs, leading to an improvement in iGMSC-mediated T cell apoptosis and therapeutic efficacy in the treatment in colitis mice. CONCLUSIONS: This study indicates that the deficient immunomodulatory function of iGMSCs could be rescued by ASA pretreatment via upregulating of FasL in mice. This strategy might serve as a practical approach to rescue deficient MSC function for further therapeutic application.


Subject(s)
Aspirin/pharmacology , Fas Ligand Protein/metabolism , Immunomodulation/drug effects , Up-Regulation/drug effects , Animals , Apoptosis/drug effects , Colitis/chemically induced , Colitis/therapy , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/genetics , Gingiva/cytology , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , RNA Interference , RNA, Small Interfering/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
3.
Leuk Res ; 68: 62-69, 2018 05.
Article in English | MEDLINE | ID: mdl-29549809

ABSTRACT

In low risk MDS, increased apoptosis of erythroid progenitors mediated via CD95 (Fas) activation has been described to result in peripheral cytopenia. Blockade of the CD95 system can improve erythropoiesis in MDS. Asunercept (APG101) is a fusion protein consisting of the extracellular domain of human CD95 and the Fc domain of human IgG1 blocking the interaction between CD95 and its ligand. Here we report on results from a phase I study in 20 transfusion-dependent low and intermediate risk MDS patients treated with intravenous asunercept (EudraCT 2012-003027-37). Primary objectives were safety and tolerability as well as pharmacodynamic effects. Secondary objectives were hematologic improvement, incidence and time to leukemic progression as well as overall survival. Frequency and severity of adverse events were in range of what could be expected in a patient cohort comprising of elderly MDS patients. Two patients experienced a serious adverse event with a suspected relationship to asunercept. The incidence of disease progression was low. In the 20 patients a decrease of the transfusion need from a mean of 10,8 (±5,1) pRBCs during the 12 weeks treatment phase to a mean of 10,0 (±4,2) pRBCs at the end of the study was observed. In conclusion, asunercept was well tolerated and showed efficacy in transfusion-dependent low and intermediate risk MDS patients. Further clinical investigation is warranted, particularly in combination with erythropoiesis stimulating agents (ESAs).


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Blood Transfusion , Immunoglobulin G/adverse effects , Immunoglobulin G/therapeutic use , Myelodysplastic Syndromes/drug therapy , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/therapeutic use , fas Receptor/therapeutic use , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Disease Progression , Dose-Response Relationship, Drug , Fas Ligand Protein/antagonists & inhibitors , Female , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/pharmacology , Male , Middle Aged , Myelodysplastic Syndromes/pathology , Prospective Studies , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacology , Risk , fas Receptor/administration & dosage
4.
Mol Cancer Res ; 16(5): 767-776, 2018 05.
Article in English | MEDLINE | ID: mdl-29453321

ABSTRACT

CD95 (Fas/APO-1), a death receptor family member, activity has been linked to tumorigenicity in multiple cancers, including glioblastoma multiforme (GBM). A phase II clinical trial on relapsed glioblastoma patients demonstrated that targeted inhibition of CD95 signaling via the CD95 ligand (CD95L) binding and neutralizing Fc-fusion protein APG101 (asunercept) prolonged patient survival. Although CD95 signaling may be relevant for multiple aspects of tumor growth, the mechanism of action of APG101 in glioblastoma is not clear. APG101 action was examined by in vitro proliferation, apoptosis, and invasion assays with human and murine glioma and human microglial cells, as well as in vivo therapy studies with orthotopic gliomas and clinical data. APG101 inhibits CD95L-mediated invasion of glioma cells. APG101 treatment was effective in glioma-bearing mice, independently of the presence or absence of CD4 and CD8 T lymphocytes, which should be sensitive to CD95L. Combined with radiotherapy, APG101 demonstrated a reduction of tumor growth, fewer tumor satellites, reduced activity of matrix metalloproteinases (MMP) as well as prolonged survival of tumor-bearing mice compared with radiotherapy alone. Inhibiting rather than inducing CD95 activity is a break-of-paradigm therapeutic approach for malignant gliomas. Evidence, both in vitro and in vivo, is provided that CD95L-binding fusion protein treatment enhanced the efficacy of radiotherapy and reduced unwanted proinfiltrative effects by reducing metalloproteinase activity by directly affecting the tumor cells.Implications: APG101 (asunercept) successfully used in a controlled phase II glioblastoma trial (NCT01071837) acts anti-invasively by inhibiting matrix metalloproteinase signaling, resulting in additive effects together with radiotherapy and helping to further develop a treatment for this devastating disease. Mol Cancer Res; 16(5); 767-76. ©2018 AACR.


Subject(s)
Fas Ligand Protein/antagonists & inhibitors , Glioblastoma/radiotherapy , Immunoglobulin G/therapeutic use , Recombinant Fusion Proteins/therapeutic use , fas Receptor/therapeutic use , Animals , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunoglobulin G/pharmacology , Mice , Recombinant Fusion Proteins/pharmacology , Signal Transduction
5.
Cell Cycle ; 17(4): 506-514, 2018.
Article in English | MEDLINE | ID: mdl-29092660

ABSTRACT

Off-target effects (OTE) are an undesired side effect of RNA interference (RNAi) caused by partial complementarity between the targeting siRNA and mRNAs other than the gene to be silenced. The death receptor CD95 and its ligand CD95L contain multiple sequences that when expressed as either si- or shRNAs kill cancer cells through a defined OTE that targets critical survival genes. Death induced by survival gene elimination (DISE) is characterized by specific morphological changes such as elongated cell shapes, senescence-like enlarged cells, appearance of large intracellular vesicles, release of mitochondrial ROS followed by activation of caspase-2, and induction of a necrotic form of mitotic catastrophe. Using genome-wide shRNA lethality screens with eight different cancer cell lines, we recently identified 651 genes as critical for the survival of cancer cells. To determine whether the toxic shRNAs targeting these 651 genes contained shRNAs that kill cancer cell through DISE rather than by silencing their respective target genes, we tested all shRNAs in the TRC library derived from a subset of these genes targeting tumor suppressors (TS). We now report that only by monitoring the responses of cancer cells following expression of shRNAs derived from these putative TS it was possible to identify DISE-inducing shRNAs in five of the genes. These data indicate that DISE in general is not an undefined toxic response of cells caused by a random OTE but rather a specific cellular response with shared features that points at a specific biological function involving multiple genes in the genome.


Subject(s)
RNA Interference , RNA, Small Interfering/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Humans , Reactive Oxygen Species/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , fas Receptor/antagonists & inhibitors , fas Receptor/genetics , fas Receptor/metabolism
6.
Mol Vis ; 23: 844-852, 2017.
Article in English | MEDLINE | ID: mdl-29259391

ABSTRACT

Purpose: Previous studies have shown that melatonin (MEL) signaling is involved in the modulation of photoreceptor viability during aging. Recent work by our laboratory suggested that MEL may protect cones by modulating the Fas/FasL-caspase-3 pathway. In this study, we first investigated the presence of MEL receptors (MT1 and MT2) in 661W cells, then whether MEL can prevent H2O2-induced cell death, and last, through which pathway MEL confers protection. Methods: The mRNA and proteins of the MEL receptors were detected with quantitative PCR (q-PCR) and immunocytochemistry, respectively. To test the protective effect of MEL, 661W cells were treated with H2O2 for 2 h in the presence or absence of MEL, a MEL agonist, and an antagonist. To study the pathways involved in H2O2-mediated cell death, a Fas/FasL antagonist was used before the exposure to H2O2. Finally, Fas/FasL and caspase-3 mRNA was analyzed with q-PCR and immunocytochemistry in cells treated with H2O2 and/or MEL. Cell viability was analyzed by using Trypan Blue. Results: Both MEL receptors (MT1 and MT2) were detected at the mRNA and protein levels in 661W cells. MEL partially prevented H2O2-mediated cell death (20-25%). This effect was replicated with IIK7 (a melatonin receptor agonist) when used at a concentration of 1 µM. Preincubation with luzindole (a melatonin receptor antagonist) blocked MEL protection. Kp7-6, an antagonist of Fas/FasL, blocked cell death caused by H2O2 similarly to what was observed for MEL. Fas, FasL, and caspase-3 expression was increased in cells treated with H2O2, and this effect was prevented by MEL. Finally, MEL treatment partially prevented the activation of caspase-3 caused by H2O2. Conclusions: The results demonstrate that MEL receptors are present and functional in 661W cells. MEL can prevent photoreceptor cell death induced by H2O2 via the inhibition of the proapoptotic pathway Fas/FasL-caspase-3.


Subject(s)
Antioxidants/pharmacology , Caspase 3/metabolism , Fas Ligand Protein/antagonists & inhibitors , Hydrogen Peroxide/toxicity , Melatonin/pharmacology , Retinal Cone Photoreceptor Cells/drug effects , fas Receptor/antagonists & inhibitors , Animals , Caspase 3/genetics , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Immunohistochemistry , Mice , Microscopy, Confocal , Oxidants/toxicity , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/genetics , Receptor, Melatonin, MT2/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , fas Receptor/genetics , fas Receptor/metabolism
7.
Nat Commun ; 8(1): 1404, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29123081

ABSTRACT

Despite impressive clinical success, cancer immunotherapy based on immune checkpoint blockade remains ineffective in many patients due to tumoral resistance. Here we use the autochthonous TiRP melanoma model, which recapitulates the tumoral resistance signature observed in human melanomas. TiRP tumors resist immunotherapy based on checkpoint blockade, cancer vaccines or adoptive T-cell therapy. TiRP tumors recruit and activate tumor-specific CD8+ T cells, but these cells then undergo apoptosis. This does not occur with isogenic transplanted tumors, which are rejected after adoptive T-cell therapy. Apoptosis of tumor-infiltrating lymphocytes can be prevented by interrupting the Fas/Fas-ligand axis, and is triggered by polymorphonuclear-myeloid-derived suppressor cells, which express high levels of Fas-ligand and are enriched in TiRP tumors. Blocking Fas-ligand increases the anti-tumor efficacy of adoptive T-cell therapy in TiRP tumors, and increases the efficacy of checkpoint blockade in transplanted tumors. Therefore, tumor-infiltrating lymphocytes apoptosis is a relevant mechanism of immunotherapy resistance, which could be blocked by interfering with the Fas/Fas-ligand pathway.


Subject(s)
Immunotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Animals , Apoptosis/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Female , Humans , Immunotherapy/methods , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/pathology , Male , Melanoma, Experimental/pathology , Mice , Mice, Transgenic , Tumor Microenvironment/immunology , fas Receptor/antagonists & inhibitors , fas Receptor/immunology
8.
Elife ; 62017 10 24.
Article in English | MEDLINE | ID: mdl-29063830

ABSTRACT

Over 80% of multiple-tested siRNAs and shRNAs targeting CD95 or CD95 ligand (CD95L) induce a form of cell death characterized by simultaneous activation of multiple cell death pathways preferentially killing transformed and cancer stem cells. We now show these si/shRNAs kill cancer cells through canonical RNAi by targeting the 3'UTR of critical survival genes in a unique form of off-target effect we call DISE (death induced by survival gene elimination). Drosha and Dicer-deficient cells, devoid of most miRNAs, are hypersensitive to DISE, suggesting cellular miRNAs protect cells from this form of cell death. By testing 4666 shRNAs derived from the CD95 and CD95L mRNA sequences and an unrelated control gene, Venus, we have identified many toxic sequences - most of them located in the open reading frame of CD95L. We propose that specific toxic RNAi-active sequences present in the genome can kill cancer cells.


Subject(s)
Antineoplastic Agents/metabolism , Cell Death , Fas Ligand Protein/antagonists & inhibitors , RNA, Small Interfering/metabolism , fas Receptor/antagonists & inhibitors , Cell Line, Tumor , Cell Survival , Humans , RNA Interference
9.
Biomed Pharmacother ; 92: 316-323, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28551553

ABSTRACT

BACKGROUND: Apoptosis is a hallmark in the pathogenesis of autoimmune hepatitis (AIH). Cytokine stresses and extrinsic apoptotic pathway have been implicated in this type of hepatic injury. Pentoxifylline plays an important role in controlling inflammation and apoptosis in different autoimmune diseases. AIM: To assess the protective effect of pentoxifylline for 30days against pro-inflammatory cytokines as tumor necrosis factor-alpha (TNF-α), interferon-gamma (INF-γ) and mediators of extrinsic apoptotic pathway involving TNF receptor 1 (TNFR1) and its ligand TNF-α and Fas receptor and its ligand (FasL) in experimental autoimmune hepatitis (EAH) model. METHODS: EAH was induced by intraperitoneal injection of syngeneic liver antigen emulsified in complete Freund's adjuvant (CFA) in male C57BL/6 mice. Five groups of mice were used: two control groups; Control PBS group and Control CFA group, EAH group and two EAH+pentoxifylline treated groups in doses (100 or 200mg/kg/d, given by oral gavage). Serum transaminase, pro-inflammatory cytokines (TNF-α and interferon-γ) and hepatic caspase-8 and 3 activities were evaluated. Signs of autoimmune hepatitis were confirmed by liver histology. In addition, hepatic TNFR1, Fas and FasL mRNA expression were assayed. RESULTS: Serum transaminase levels and signs of AIH observed in EAH mice were significantly reduced by pentoxifylline. Upregulated serum TNF-α, IFN-γ, hepatic caspase-8 and 3 activities and TNFR1, Fas and FasL mRNA expression in liver tissues in EAH group were significantly downregulated by pentoxifylline. CONCLUSION: Pentoxifylline protects against syngeneic liver antigen induced hepatitis and associating apoptosis through attenuating the exaggerated cytokine release and extrinsic apoptotic pathway. Thus, this may represent a new therapeutic strategy for hepatitis.


Subject(s)
Cytokines/antagonists & inhibitors , Fas Ligand Protein/antagonists & inhibitors , Hepatitis, Autoimmune/drug therapy , Oxidative Stress/drug effects , Pentoxifylline/therapeutic use , fas Receptor/antagonists & inhibitors , Animals , Cytokines/metabolism , Fas Ligand Protein/metabolism , Hepatitis, Autoimmune/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/physiology , Pentoxifylline/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , fas Receptor/metabolism
10.
Nat Rev Rheumatol ; 13(4): 217-233, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28275260

ABSTRACT

TNF blockers are highly efficacious at dampening inflammation and reducing symptoms in rheumatic diseases such as rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis, and also in nonrheumatic syndromes such as inflammatory bowel disease. As TNF belongs to a superfamily of 19 structurally related proteins that have both proinflammatory and anti-inflammatory activity, reagents that disrupt the interaction between proinflammatory TNF family cytokines and their receptors, or agonize the anti-inflammatory receptors, are being considered for the treatment of rheumatic diseases. Biologic agents that block B cell activating factor (BAFF) and receptor activator of nuclear factor-κB ligand (RANKL) have been approved for the treatment of systemic lupus erythematosus and osteoporosis, respectively. In this Review, we focus on additional members of the TNF superfamily that could be relevant for the pathogenesis of rheumatic disease, including those that can strongly promote activity of immune cells or increase activity of tissue cells, as well as those that promote death pathways and might limit inflammation. We examine preclinical mouse and human data linking these molecules to the control of damage in the joints, muscle, bone or other tissues, and discuss their potential as targets for future therapy of rheumatic diseases.


Subject(s)
Molecular Targeted Therapy , Rheumatic Diseases/drug therapy , Rheumatic Diseases/immunology , Tumor Necrosis Factor Inhibitors , Tumor Necrosis Factors/metabolism , 4-1BB Ligand/antagonists & inhibitors , 4-1BB Ligand/metabolism , Animals , CD27 Ligand/antagonists & inhibitors , CD27 Ligand/metabolism , CD40 Ligand/antagonists & inhibitors , CD40 Ligand/metabolism , Cell Death , Cytokine TWEAK , Dendritic Cells/immunology , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/metabolism , Humans , Immune Tolerance , Lymphocyte Activation , Lymphotoxin-alpha/antagonists & inhibitors , Lymphotoxin-alpha/metabolism , OX40 Ligand/antagonists & inhibitors , OX40 Ligand/metabolism , Signal Transduction , T-Lymphocytes/immunology , TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/antagonists & inhibitors , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/antagonists & inhibitors , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Tumor Necrosis Factors/immunology
11.
Int J Mol Sci ; 17(11)2016 Nov 23.
Article in English | MEDLINE | ID: mdl-27886059

ABSTRACT

Our previous study has reported the anti-tumor effect of oleandrin on osteosarcoma (OS) cells. In the current study, we mainly explored its potential regulation on intrinsic and extrinsic apoptotic pathway in OS cells. Cells apoptosis, reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were detected using fluorescence staining and flow cytometry. Caspase-3 activity was detected using a commercial kit. The levels of cytoplasmic cytochrome c, mitochondrial cytochrome c, bcl-2, bax, caspase-9, Fas, FasL, caspase-8 and caspase-3 were detected by Western blotting. z-VAD-fmk was applied to block both intrinsic and extrinsic apoptosis pathways, and cells apoptosis was also tested. Furthermore, we used z-LEHD-fmk and Fas blocking antibody to inhibit intrinsic and extrinsic pathways, separately, and the selectivity of oleandrin on these pathways was explored. Results showed that oleandrin induced the apoptosis of OS cells, which was accompanied by an increase in ROS and a decrease in MMP. Furthermore, cytochrome c level was reduced in mitochondria but elevated in the cytoplasm. Caspase-3 activity was enhanced by oleandrin in a concentration- and time-dependent manner. Oleandrin also down-regulated the expression of bcl-2, but up-regulated bax, caspase-9, Fas, FasL, caspase-8 and caspase-3. In addition, the suppression of both apoptotic pathways by z-VAD-fmk greatly reverted the oleandrin-induced apoptosis. Moreover, the suppression of one pathway by a corresponding inhibitor did not affect the regulation of oleandrin on another pathway. Taken together, we concluded that oleandrin induced apoptosis of OS cells via activating both intrinsic and extrinsic apoptotic pathways.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cardenolides/pharmacology , Cardiac Glycosides/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Osteoblasts/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Antibodies, Neutralizing/pharmacology , Caspase 3/genetics , Caspase 3/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Tumor , Cytochromes c/genetics , Cytochromes c/metabolism , Fas Ligand Protein/agonists , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Oligopeptides/pharmacology , Osteoblasts/metabolism , Osteoblasts/pathology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Signal Transduction , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
12.
Arch Oral Biol ; 71: 31-37, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27421097

ABSTRACT

OBJECTIVE: High fluoride can induce stress-mediated apoptosis and degradation of ameloblasts. Fas ligand (FasL) has been regarded as a key regulator in intracellular responses for stress-induced apoptosis in reproductive or cancerous cell lineages. The objective of this study is to explore the role of FasL in the regulation of ameloblast ultrastructure damage. DESIGN: Primary ameloblasts were isolated from the molar tooth germ of 4-day-old SD rats. The ameloblasts were incubated with 3.2mM NaF or nothing. After incubation for different time arranging from 12h to 72h, ELISA was used to detected the secretion levels of FasL in the medium. Then at 48h post treatment, the ameloblast ultrastructure was detected with Transmission Electron Microscopy (TEM) and Scanning Electron Microscopy (SEM), and expression of apoptotic proteins and peroxidative enzymes/products were examined. Finally, a specific FasL inhibitor was applied to co-treat the ameloblasts with NaF, and the ameloblast ultrastructure was detected with TEM and SEM. RESULTS: The secretion of FasL was notably increased by 3.2mM NaF treatment, and the increase reached to the peak after incubation for 48h. High fluoride incubation damaged the ameloblast untrastructure manifesting a series of intracelluar stress responsing cell organelle destruction, and a marked increase in expression of apoptotic genes and oxidative stress. The FasL inhibitor treatment partially mitigated the untrastructure damage caused by high dose NaF. CONLUSION: High-fluoride leads to damage of the ameloblast ultrastructure through paritially acitivating the FasL signalling pathway.


Subject(s)
Ameloblasts/drug effects , Ameloblasts/ultrastructure , Fas Ligand Protein/metabolism , Signal Transduction/drug effects , Sodium Fluoride/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fas Ligand Protein/antagonists & inhibitors , Microscopy, Electron , Molar , Oxidative Stress , Rats , Tooth Germ/drug effects
13.
Cell Death Dis ; 7: e2232, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27195678

ABSTRACT

Chagas disease is caused by infection with the protozoan Trypanosoma cruzi. CD8 T-lymphocytes help to control infection, but apoptosis of CD8 T cells disrupts immunity and efferocytosis can enhance parasite infection within macrophages. Here, we investigate how apoptosis of activated CD8 T cells affects M1 and M2 macrophage phenotypes. First, we found that CD8 T-lymphocytes and inflammatory monocytes/macrophages infiltrate peritoneum during acute T. cruzi infection. We show that treatment with anti-Fas ligand (FasL) prevents lymphocyte apoptosis, upregulates type-1 responses to parasite antigens, and reduces infection in macrophages cocultured with activated CD8 T cells. Anti-FasL skews mixed M1/M2 macrophage profiles into polarized M1 phenotype, both in vitro and following injection in infected mice. Moreover, inhibition of T-cell apoptosis induces a broad reprogramming of cytokine responses and improves macrophage-mediated immunity to T. cruzi. The results indicate that disposal of apoptotic CD8 T cells increases M2-macrophage differentiation and contributes to parasite persistence.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chagas Disease/immunology , Fas Ligand Protein/antagonists & inhibitors , Host-Parasite Interactions , Immunity, Cellular/drug effects , Macrophages/immunology , Animals , Antibodies, Neutralizing/pharmacology , Apoptosis/drug effects , Asialoglycoproteins/genetics , Asialoglycoproteins/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/parasitology , Cell Communication/drug effects , Cell Movement/drug effects , Chagas Disease/drug therapy , Chagas Disease/genetics , Chagas Disease/parasitology , Coculture Techniques , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Gene Expression Regulation , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p35/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymphocyte Activation , Macrophages/drug effects , Macrophages/parasitology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Phenotype , Trypanosoma cruzi/growth & development , Trypanosoma cruzi/immunology
14.
Discov Med ; 21(115): 213-9, 2016 03.
Article in English | MEDLINE | ID: mdl-27115172

ABSTRACT

Immunotherapy has revolutionized treatment of cancers and autoimmune diseases. Bucking the trend, however, is type 1 diabetes (T1D), although it is one of best understood autoimmune diseases and individuals at genetic risk are identifiable with high certainty. Here we review the major obstacles associated with pan-B-cell-depletion using rituximab (RTX) and discuss the notion that B cell-directed therapy may be most effective as a preventive measure. We suggest that it will be more productive to aim at identifying and targeting autoreactive B cells rather than making adjustments to pan-B cell depletion and that non-conventional alternative therapies such as antibody blockade of FasL to bolster IL-10-producing Breg cells, which work successfully in mice, should be considered.


Subject(s)
B-Lymphocytes/drug effects , Diabetes Mellitus, Type 1/therapy , Immunologic Factors/therapeutic use , Immunotherapy/methods , Rituximab/therapeutic use , Animals , Antigens, CD20/immunology , Antigens, CD20/metabolism , Autoimmunity/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Clinical Trials as Topic , Drug Therapy, Combination , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/metabolism , Humans , Immunologic Factors/adverse effects , Immunotherapy/adverse effects , Interleukin-10/metabolism , Islets of Langerhans/immunology , Islets of Langerhans Transplantation/immunology , Mice , Rituximab/adverse effects , Treatment Outcome
15.
Brain Res ; 1637: 64-70, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26892027

ABSTRACT

Respecting the selective inhibition of peptides on protein-protein interactions, they might become potent methods in ischemic stroke therapy. In this study, we investigated the effect of PDZ1 inhibitor peptide on ischemic neuron apoptosis and the relative mechanism. Results showed that PDZ1 inhibitor peptide, which significantly disrupted GluK2-PSD-95 interaction, efficiently protected neuron from ischemia/reperfusion-induced apoptosis. Further, PDZ1 inhibited FasL expression, DISC assembly and activation of Caspase 8, Bid, Caspase 9 and Caspase 3 after global brain ischemia. Based on our previous report that GluK2-PSD-95 pathway increased FasL expression after global brain ischemia, the neuron protection effect of PDZ1 inhibitor peptide was considered to be achieved by disrupting GluK2-PSD-95 interaction and subsequently inhibiting FasL expression and Fas apoptosis pathway.


Subject(s)
Brain Ischemia/drug therapy , Guanylate Kinases/antagonists & inhibitors , Peptides/pharmacology , Receptors, Kainic Acid/antagonists & inhibitors , Animals , Apoptosis/drug effects , Brain Ischemia/metabolism , Brain Ischemia/pathology , Caspases/metabolism , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/metabolism , Guanylate Kinases/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/metabolism , PDZ Domains , Rats , Rats, Sprague-Dawley , Receptors, Kainic Acid/metabolism , Reperfusion Injury/metabolism , Signal Transduction , GluK2 Kainate Receptor
16.
J Biol Chem ; 291(3): 1221-34, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26582200

ABSTRACT

Death receptors are members of the tumor necrosis factor receptor superfamily involved in the extrinsic apoptotic pathway. Lifeguard (LFG) is a death receptor antagonist mainly expressed in the nervous system that specifically blocks Fas ligand (FasL)-induced apoptosis. To investigate its mechanism of action, we studied its subcellular localization and its interaction with members of the Bcl-2 family proteins. We performed an analysis of LFG subcellular localization in murine cortical neurons and found that LFG localizes mainly to the ER and Golgi. We confirmed these results with subcellular fractionation experiments. Moreover, we show by co-immunoprecipitation experiments that LFG interacts with Bcl-XL and Bcl-2, but not with Bax or Bak, and this interaction likely occurs in the endoplasmic reticulum. We further investigated the relationship between LFG and Bcl-XL in the inhibition of apoptosis and found that LFG protects only type II apoptotic cells from FasL-induced death in a Bcl-XL dependent manner. The observation that LFG itself is not located in mitochondria raises the question as to whether LFG in the ER participates in FasL-induced death. Indeed, we investigated the degree of calcium mobilization after FasL stimulation and found that LFG inhibits calcium release from the ER, a process that correlates with LFG blockage of cytochrome c release to the cytosol and caspase activation. On the basis of our observations, we propose that there is a required step in the induction of type II apoptotic cell death that involves calcium mobilization from the ER and that this step is modulated by LFG.


Subject(s)
Apoptosis , Calcium Signaling , Endoplasmic Reticulum/metabolism , Fas Ligand Protein/antagonists & inhibitors , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Cell Line , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Female , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice, Inbred C57BL , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/cytology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Transport , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
17.
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
18.
Toxicol Ind Health ; 32(9): 1628-38, 2016 Sep.
Article in English | MEDLINE | ID: mdl-25757482

ABSTRACT

OBJECTIVE: To investigate the contributions and underlying molecular mechanisms of annexin A5 toward silica-induced pulmonary fibrosis. METHODS: Male C57BL/6 mice were randomly divided into three groups and instilled intratracheally with silica, saline, or air. Mice were euthanized at 3, 7, 14, or 28 days following treatment. Annexin A5 levels in serum and lung tissues were detected by enzyme-linked immunosorbant assay (ELISA) assays or Western blots. The association of annexin A5 levels with silica-induced lung fibrosis was further investigated in the macrophage cell line, RAW264.7. Following exposure of these cells to silica at a concentration of 200 µg/ml for 6 or 12 h, the expression levels of transforming growth factor ß1 (TGF-ß1), interleukin 1α (IL-1α), Fas ligand (FasL), and their downstream targets were evaluated by Western blots. Furthermore, annexin A5 and FasL were knocked down by small interfering ribonucleic acid (siRNA) and TGF-ß1 secretion into the cell culture medium was measured by ELISA assays or Western blots. RESULTS: Mice treated with silica demonstrated lung fibrosis at 28 days following exposure, whereas, in controls, only mild and transient inflammation was evident at day 3 and day 7 postinstillation and was not present at day 14. Furthermore, silica-exposed mice exhibited significantly (p < 0.05) elevated levels of annexin A5 in serum and lung tissues, relative to control groups. Consistent with these findings, silica exposure of RAW264.7 cells for 6 or 12 h, led to an annexin A5-dependent increase in the expression levels of TGF-ß1, IL-1α, FasL, and their downstream target molecules. These silica-induced changes were reversed by siRNA-mediated knockdown of annexin A5, but downregulation of FasL led to increased annexin A5 expression and reduced levels of TGF-ß1, IL-1α, and FasL downstream target molecules. CONCLUSIONS: These findings define a role of annexin A5 in promoting macrophage activation via Fas/FasL pathways in silica-induced lung fibrosis.


Subject(s)
Annexin A5/metabolism , Disease Models, Animal , Macrophage Activation/drug effects , Pulmonary Fibrosis/etiology , Respiratory Mucosa/drug effects , Silicon Dioxide/toxicity , Silicosis/physiopathology , Air Pollutants/chemistry , Air Pollutants/toxicity , Animals , Annexin A5/antagonists & inhibitors , Annexin A5/blood , Annexin A5/genetics , Cytokines/agonists , Cytokines/metabolism , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Particulate Matter/chemistry , Particulate Matter/toxicity , RAW 264.7 Cells , RNA Interference , Random Allocation , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Signal Transduction/drug effects , Silicon Dioxide/chemistry , Silicosis/immunology , Silicosis/metabolism , Silicosis/pathology , Specific Pathogen-Free Organisms
19.
Viruses ; 7(8): 4756-71, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26295407

ABSTRACT

Baicalin is a flavonoid compound extracted from Scutellaria roots that has been reported to possess antibacterial, anti-inflammatory, and antiviral activities. However, the antiviral effect of baicalin on enterovirus 71 (EV71) is still unknown. In this study, we found that baicalin showed inhibitory activity on EV71 infection and was independent of direct virucidal or prophylactic effect and inhibitory viral absorption. The expressions of EV71/3D mRNA and polymerase were significantly blocked by baicalin treatment at early stages of EV71 infection. In addition, baicalin could decrease the expressions of FasL and caspase-3, as well as inhibit the apoptosis of EV71-infected human embryonal rhabdomyosarcoma (RD) cells. Altogether, these results indicate that baicalin exhibits potent antiviral effect on EV71 infection, probably through inhibiting EV71/3D polymerase expression and Fas/FasL signaling pathways.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Flavonoids/pharmacology , Cell Line , DNA-Directed RNA Polymerases/antagonists & inhibitors , Fas Ligand Protein/antagonists & inhibitors , Flavonoids/isolation & purification , Humans , Microbial Sensitivity Tests , Scutellaria/chemistry , Signal Transduction/drug effects
20.
Biochem Biophys Res Commun ; 463(4): 1144-51, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26102031

ABSTRACT

Resistance to Fas Ligand (FasL) mediated apoptosis plays an important role in tumorigenesis. Decoy receptor 3 (DcR3) is reported to interact with FasL and is overexpressed in some malignant tumors. We sought to investigate the role of DcR3 in resistance to FasL in pancreatic cancer. We compared expression of apoptosis related genes between FasL-resistant SW1990 and FasL-sensitive Patu8988 pancreatic cell lines by microarray analysis. We explored the impact of siRNA knockdown of, or exogenous supplementation with, DcR3 on FasL-induced cell growth inhibition in pancreatic cancer cell lines and expression of proteins involved in apoptotic signaling. We assessed the level of DcR3 protein and ERK1/2 phosphorylation in tumor and non-tumor tissue samples of 66 patients with pancreatic carcinoma. RNAi knockdown of DcR3 expression in SW1990 cells reduced resistance to FasL-induced apoptosis, and supplementation of Patu8988 with rDcR3 had the opposite effect. RNAi knockdown of DcR3 in SW1990 cells elevated expression of caspase 3, 8 and 9, and reduced ERK1/2 phosphorylation (P < 0.05), but did not alter phosphorylated-Akt expression. 47 tumor tissue specimens, but only 15 matched non-tumor specimens stained for DcR3 (χ(2) = 31.1447, P < 0.001). The proliferation index of DcR3 positive specimens (14.26  ±  2.67%) was significantly higher than that of DcR3 negative specimens (43.58  ±  7.88%, P < 0.01). DcR3 expression positively correlated with p-ERK1/2 expression in pancreatic cancer tissues (r = 0.607, P < 0.001). DcR3 enhances ERK1/2 phosphorylation and opposes FasL signaling in pancreatic cancer cells.


Subject(s)
Apoptosis/physiology , Fas Ligand Protein/antagonists & inhibitors , MAP Kinase Signaling System , Pancreatic Neoplasms/pathology , Receptors, Tumor Necrosis Factor, Member 6b/physiology , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Fas Ligand Protein/physiology , Humans , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/metabolism , Phosphorylation , RNA Interference , Receptors, Tumor Necrosis Factor, Member 6b/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...