Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Oncotarget ; 7(51): 84388-84397, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27768597

ABSTRACT

γδ T cells has been shown to exhibit profound antitumor effects in a broad range of tumor entities, including OS. However, resistance to γδ T cells is a serious problem in the management of OS. This study investigates the impact of celastrol on the expression of death receptors 4/5 (DR4/5) on OS cell lines (HOS, U2OS) and cancer cell lysis by γδ T cells. The results showed that celastrol increased transcription of DR4/5 in HOS and U2OS, leading to increased cell surface, and total DR4/5 protein expression. Celastrol sensitizes OS cell lines or autologous OS cells to healthy donors-derived or OS patient-derived γδ T cell cytotoxicity in vitro. The induction of DR4/5 molecules increased lysis of HOS and U2OS by γδ T cells which was abolished by addition of a blocking TRAIL antibody. Importantly, the cytotoxic activity of γδ T cells was unaltered by small-dose celastrol. Taken together, our data show that celastrol up-regulated DR4/5 on OS cells to be responsible for intercellular TRAIL/APO-2L crosslink that confers increased cancer cell lysis by γδ T cells. These results suggest the clinical evaluation of celastrol in OS, especially in combination with immunotherapy approaches employing adoptive γδ T cell transfer.


Subject(s)
Cytotoxicity, Immunologic/drug effects , Osteosarcoma/immunology , Receptors, Death Domain/immunology , T-Lymphocytes/immunology , Triterpenes/pharmacology , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic/immunology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Osteosarcoma/genetics , Osteosarcoma/metabolism , Pentacyclic Triterpenes , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Death Domain/genetics , Receptors, Death Domain/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , T-Lymphocytes/metabolism , Up-Regulation/drug effects
2.
PLoS One ; 10(8): e0136209, 2015.
Article in English | MEDLINE | ID: mdl-26295571

ABSTRACT

OBJECTIVE: The aim of this study was to explore the therapeutic effect of natural killer (NK) cells on human doxorubicin-sensitive and resistant breast adenocarcinoma. METHODS: Human doxorubicin-sensitive and resistant breast cancer cell lines (MCF-7 and MCF-7/ADR) were tagged with renilla luciferase (Rluc) (MCF-7/RC and MCF-7/ADR/RC). NK cells were tagged with enhanced firefly luciferase (effluc) using a recombinant retrovirus transfection (NKF). Expression of Rluc, effluc, and NK cell surface markers CD16, CD56 as well as death receptors, DR4 and DR5, were assessed by using flow cytometry. In vitro cytotoxic effect of NK to MCF-7 and MCF-7/ADR was measured and in vivo bioluminescence imaging was also performed to visualize MCF-7/RC, MCF-7/ADR, and NKF in an animal model. RESULTS: NK92-MI, MCF-7, and MCF-7/ADR cells were successfully labeled with Rluc or effluc. Both the target breast cancer cells (with Rluc) and therapeutic NK cells (with effluc) were noninvasively visualized in nude mice. Doxorubicin-resistant breast cancer cells (MCF-7/ADR) presented a higher expression of DR5 and were more sensitive to NK cells compared with doxorubicin-sensitive breast cancer cells (MCF-7). CONCLUSION: The results of present study suggest that NK cell therapy has a therapeutic effect on doxorubicin-sensitive and resistant breast cancer cells.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/therapy , Breast/pathology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Animals , Breast/drug effects , Breast/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell- and Tissue-Based Therapy , Female , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Optical Imaging , Receptors, Death Domain/analysis , Receptors, Death Domain/immunology
3.
Trends Pharmacol Sci ; 35(10): 520-36, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25128958

ABSTRACT

Selective killing of cancer cells is one of the major goals of cancer therapy. Although chemotherapeutic agents are being used for cancer treatment, they lack selectivity toward tumor cells. Among the six different death receptors (DRs) identified to date, DR4 and DR5 are selectively expressed on cancer cells. Therefore, unlike chemotherapeutic agents, these receptors can potentially mediate selective killing of tumor cells. In this review we outline various nutraceuticals derived from 'Mother Nature' that can upregulate DRs and thus potentiate apoptosis. These nutraceuticals increase tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL)-induced apoptosis of cancer cells through different mechanisms. First, nutraceuticals have been found to induce DRs through the upregulation of various signaling molecules. Second, nutraceuticals can downregulate tumor cell-survival pathways. Third, nutraceuticals alone have been found to activate cell-death pathways. Although both TRAIL and agonistic antibodies against DR4 and DR5 are in clinical trials, combination with nutraceuticals is likely to boost their anticancer potential.


Subject(s)
Antineoplastic Agents/pharmacology , Dietary Supplements , Receptors, Death Domain/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Antibodies/immunology , Antibodies/pharmacology , Drug Synergism , Humans , Receptors, Death Domain/agonists , Receptors, Death Domain/immunology
4.
Cytokine Growth Factor Rev ; 25(2): 235-43, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24440054

ABSTRACT

Gastrointestinal bacterial pathogens such as enteropathogenic Escherichia coli, Salmonella and Shigella control inflammatory and apoptotic signaling in human intestinal cells to establish infection, replicate and disseminate to other hosts. These pathogens manipulate host cell signaling through the translocation of virulence effector proteins directly into the host cell cytoplasm, which then target various signaling pathways. Death receptors such as TNFR1, FAS and TRAIL-R induce signaling cascades that are crucial to the clearance of pathogens, and as such are major targets for inhibition by pathogens. This review focuses on what is known about how bacterial gut pathogens inhibit death receptor signaling to suppress inflammation and prevent apoptosis.


Subject(s)
Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Receptors, Death Domain/antagonists & inhibitors , Receptors, Death Domain/immunology , Apoptosis/immunology , Bacterial Secretion Systems/immunology , Escherichia coli/immunology , Humans , Inflammation/immunology , Microbiota/immunology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Salmonella/immunology , Shigella/immunology , Signal Transduction/immunology
5.
J Virol ; 88(2): 1080-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24198425

ABSTRACT

Apoptosis is an important mechanism of West Nile virus (WNV) pathogenesis within the central nervous system (CNS). The signaling pathways that result in WNV-induced apoptotic neuronal death within the CNS have not been established. In this study, we identified death receptor (DR)-induced apoptosis as a pathway that may be important in WNV pathogenesis, based on the pattern of differential gene expression in WNV-infected, compared to uninfected, brains. Reverse transcription-PCR (RT-PCR) and Western blotting confirmed that genes involved in DR-induced apoptotic signaling are upregulated in the brain following WNV infection. Activity of the DR-associated initiator caspase, caspase 8, was also increased in the brains of WNV-infected mice and occurred in association with cleavage of Bid and activation of caspase 9. These results demonstrate that DR-induced apoptotic signaling is activated in the brain following WNV infection and suggest that the caspase 8-dependent cleavage of Bid promotes intrinsic apoptotic signaling within the brains of infected animals. Utilization of a novel ex vivo brain slice culture (BSC) model of WNV encephalitis revealed that inhibition of caspase 8 decreases virus-induced activation of caspase 3 and tissue injury. The BSC model allows us to examine WNV-induced pathogenesis in the absence of a peripheral immune response. Thus, our results indicate that WNV-induced neuronal injury in the brain is mediated by DR-induced apoptosis signaling and can occur in the absence of infiltrating immune cells. However, astrocytes and microglia were activated in WNV-infected BSC, suggesting that local immune responses influence WNV pathogenesis.


Subject(s)
Apoptosis , Brain/cytology , Peripheral Nervous System/immunology , Receptors, Death Domain/immunology , West Nile Fever/physiopathology , West Nile virus/physiology , Animals , Brain/enzymology , Brain/immunology , Brain/virology , Caspase 3/genetics , Caspase 3/immunology , Caspase 8/genetics , Caspase 8/immunology , Caspase 9/genetics , Caspase 9/immunology , Female , Humans , Male , Mice , Peripheral Nervous System/enzymology , Peripheral Nervous System/virology , Receptors, Death Domain/genetics , Signal Transduction , West Nile Fever/enzymology , West Nile Fever/genetics , West Nile Fever/immunology , West Nile virus/genetics
6.
Anal Chem ; 85(11): 5609-16, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23621478

ABSTRACT

The variable susceptibility to the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) treatment observed in various types of leukemia cells is related to the difference in the expression levels of death receptors, DR4 and DR5, on the cell surfaces. Quantifying the DR4/DR5 expression status on leukemia cell surfaces is of vital importance to the development of diagnostic tools to guide death receptor-based leukemia treatment. Taking the full advantages of novel nanobiotechnology, we have developed a robust electrochemical cytosensing approach toward ultrasensitive detection of leukemia cells with detection limit as low as ~40 cells and quantitative evaluation of DR4/DR5 expression on leukemia cell surfaces. The optimization of electron transfer and cell capture processes at specifically tailored nanobiointerfaces and the incorporation of multiple functions into rationally designed nanoprobes provide unique opportunities of integrating high specificity and signal amplification on one electrochemical cytosensor. The high sensitivity and selectivity of this electrochemical cytosensing approach also allows us to evaluate the dynamic alteration of DR4/DR5 expression on the surfaces of living cells in response to drug treatments. Using the TRAIL-resistant HL-60 cells and TRAIL-sensitive Jurkat cells as model cells, we have further verified that the TRAIL susceptibility of various types of leukemia cells is directly correlated to the surface expression levels of DR4/DR5. This versatile electrochemical cytosensing platform is believed to be of great clinical value for the early diagnosis of human leukemia and the evaluation of therapeutic effects on leukemia patients after radiation therapy or drug treatment.


Subject(s)
Biosensing Techniques/methods , Electrochemical Techniques/methods , Gold/chemistry , Leukemia/diagnosis , Metal Nanoparticles/chemistry , Receptors, Death Domain/metabolism , Antioxidants/pharmacology , Carbon/chemistry , Electrodes , Ferric Compounds/metabolism , Flow Cytometry , HL-60 Cells , Horseradish Peroxidase/metabolism , Humans , Immunoassay , Leukemia/drug therapy , Leukemia/metabolism , Melatonin/pharmacology , Nitrogen/chemistry , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Receptors, Death Domain/immunology , TNF-Related Apoptosis-Inducing Ligand/metabolism
7.
Cancer Lett ; 332(2): 175-83, 2013 May 28.
Article in English | MEDLINE | ID: mdl-21215513

ABSTRACT

Ideally, an immunotoxin should be inactive 'en route', acquire activity only after tumor cell surface binding and have no off-target effects towards normal cells. In this respect, antibody-based fusion proteins that exploit the tumor-selective pro-apoptotic death ligands sFasL and sTRAIL appear promising. Soluble FasL largely lacks receptor-activating potential, whereas sTRAIL is inactive towards normal cells. Fusion proteins in which an anti-tumor antibody fragment (scFv) is fused to sFasL or sTRAIL prove to be essentially inactive when soluble, while gaining potent anti-tumor activity after selective binding to a predefined tumor-associated cell surface antigen. Importantly, off-target binding by scFv:sTRAIL to normal cells showed no signs of toxicity. In this review, we highlight the rationale and perspectives of scFv:TRAIL/scFv:sFasL based fusion proteins for cancer therapy.


Subject(s)
Antibodies/immunology , Neoplasms/immunology , Receptors, Death Domain/immunology , Recombinant Fusion Proteins/pharmacology , Animals , Apoptosis , Cell Membrane/metabolism , Disease Models, Animal , Fas Ligand Protein/metabolism , Female , Humans , Immunotherapy/methods , Ligands , Male , Neoplasms/drug therapy , Signal Transduction
8.
Adv Exp Med Biol ; 738: 124-43, 2012.
Article in English | MEDLINE | ID: mdl-22399377

ABSTRACT

Programmed Cell Death is essential for the life cycle of many organisms. Cell death in multicellular organisms can occur as a consequence of massive damage (necrosis) or in a controlled form, through engagement of diverse biochemical programs. The best well known form of programmed cell death is apoptosis. Apoptosis occurs in animals as a consequence of a variety of stimuli including stress and social signals and it plays essential roles in morphogenesis and immune defense. The machinery of apoptosis is well conserved among animals and it is composed of caspases (the proteases which execute cell death), adapter proteins (caspase activators), Bcl-2 family proteins and Inhibitor of Apoptosis Proteins (IAPs). We will describe in this chapter the main apoptotic pathways in animals: the extrinsic (death receptor-mediated), the intrinsic/mitochondrial and the Granzyme B pathway. Other forms of non-apoptotic Programmed Cell Death which occur in animals will also be discussed. We will summarize the current knowledge about apoptotic-like and other forms of cell death in other organisms such as plants and protists.Additionally, we will discuss the hypothesis that apoptosis originated as part of a host defense mechanism. We will explore the similarities between the protein complexes which mediate apoptosis (apoptosomes) and complexes involved in immunity: inflammasomes. Additional functions of apoptotic proteins related to immune function will be summarized, in an effort to explore the evolutionary origins of cell death.


Subject(s)
Apoptosis/immunology , Biological Evolution , Signal Transduction/immunology , Stress, Physiological/immunology , Animals , Granzymes/immunology , Humans , Inhibitor of Apoptosis Proteins/immunology , Mitochondria/immunology , Receptors, Death Domain/immunology
9.
Int J Oncol ; 40(3): 645-54, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22076446

ABSTRACT

We characterized Fas immunoreactivity, functionality and its role in the response to mitomycin-C (MMC) chemotherapy in vitro in cell lines and in vivo in bladder washings from 23 transitional cell carcinoma of the bladder (TCCB) patients, harvested prior to and during MMC intravesical treatment. Having established the importance of functional Fas, we investigated the methylation and exon 9 mutation as mechanisms of Fas silencing in TCCB. For the first time, we report p53 up-regulation in 9/14 and Fas up-regulation in 7/9 TCCB patients during intravesical MMC treatment. Fas immunoreactivity was strong in the TCCB cell line T24 and in 17/20 (85%) tumor samples from patients with advanced TCCB. T24 and HT1376 cells were resistant to MMC and recombinant Fas ligand, whilst RT4 cells were responsive to Fas ligand and MMC. Using RT4 cells as a model, siRNA targeting p53 significantly reduced MMC-induced p53 and Fas up-regulation and stable DN-FADD transfection decreased MMC-induced apoptosis, suggesting that functional Fas enhances chemotherapy responses in a p53-dependent manner. In HT1376 cells, 5-aza-2-deoxycytidine (12 µM) induced Fas immunoreactivity and reversed methylation at CpG site -548 within the Fas promoter. This site was methylated in 13/24 (54%) TCCB patient samples assessed using Methylation-Specific Polymerase Chain Reaction. There was no methylation at either the p53 enhancer region within the first intron or at the SP-1 binding region in the promoter and no mutation within exon 9 in tumor DNA extracted from 38 patients. Methylation at CpG site -548 is a potential target for demethylating drugs.


Subject(s)
Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/immunology , DNA Methylation , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , fas Receptor/genetics , fas Receptor/immunology , Aged , Apoptosis/genetics , Apoptosis/immunology , Caco-2 Cells , Cell Line, Tumor , CpG Islands , Drug Resistance, Neoplasm , Exons , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Female , Follow-Up Studies , Gene Silencing , Genes, p53 , Humans , Introns , Male , Mitomycin/therapeutic use , Mutation , Promoter Regions, Genetic , Receptors, Death Domain/genetics , Receptors, Death Domain/immunology , Transfection/methods , Up-Regulation
10.
Clin Dev Immunol ; 2011: 439752, 2011.
Article in English | MEDLINE | ID: mdl-22162711

ABSTRACT

Stereotactic radiation approaches are gaining more popularity for the treatment of intracranial as well as extracranial tumors in organs such as the liver and lung. Technology, rather than biology, is driving the rapid adoption of stereotactic body radiation therapy (SBRT), also known as stereotactic ablative radiotherapy (SABR), in the clinic due to advances in precise positioning and targeting. Dramatic improvements in tumor control have been demonstrated; however, our knowledge of normal tissue biology response mechanisms to large fraction sizes is lacking. Herein, we will discuss how SABR can induce cellular expression of MHC I, adhesion molecules, costimulatory molecules, heat shock proteins, inflammatory mediators, immunomodulatory cytokines, and death receptors to enhance antitumor immune responses.


Subject(s)
Immune System/radiation effects , Neoplasms/immunology , Neoplasms/surgery , Radiosurgery/methods , Animals , Apoptosis/immunology , B7-1 Antigen/immunology , Brain Neoplasms/immunology , Brain Neoplasms/surgery , Cell Adhesion Molecules/immunology , Cytokines/immunology , Heat-Shock Proteins/immunology , Humans , Immune Tolerance , Liver Neoplasms/immunology , Liver Neoplasms/surgery , Lung Neoplasms/immunology , Lung Neoplasms/surgery , Major Histocompatibility Complex/immunology , Mice , Mice, Knockout , Receptors, Death Domain/immunology , Tumor Escape
11.
Trends Immunol ; 31(7): 270-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20579936

ABSTRACT

Plasmacytoid dendritic cells (PDCs) represent a subset of circulating leukocytes characterized by the ability to release high levels of type I interferon (IFN). Under homeostatic conditions PDCs are confined to primary and secondary lymphoid organs. This is consistent with the restricted profile of functional chemotactic receptors expressed by circulating PDCs (i.e. CXCR4 and ChemR23). Accumulation of PDCs in non-lymphoid tissue is, however, observed in certain autoimmune diseases, allergic reactions and tumors. Indeed, PDCs are now considered to be involved in the pathogenesis of diseases characterized by a type I IFN-signature and are considered as a promising target for new intervention strategies. Here, current knowledge of the molecular mechanisms involved in the recruitment of PDCs under homeostatic and pathological conditions are summarized.


Subject(s)
Cell Movement , Dendritic Cells/cytology , Dendritic Cells/immunology , Animals , Humans , Immunity, Innate , Receptors, Death Domain/immunology , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology
13.
Oncogene ; 29(11): 1641-52, 2010 Mar 18.
Article in English | MEDLINE | ID: mdl-19966861

ABSTRACT

Most cancer cells exhibit increased glycolysis for generation of their energy supply. This specificity could be used to preferentially kill these cells. In this study, we identified the signaling pathway initiated by glycolysis inhibition that results in sensitization to death receptor (DR)-induced apoptosis. We showed, in several human cancer cell lines (such as Jurkat, HeLa, U937), that glucose removal or the use of nonmetabolizable form of glucose (2-deoxyglucose) dramatically enhances apoptosis induced by Fas or by tumor necrosis factor-related apoptosis-inducing ligand. This sensitization is controlled through the adenosine monophosphate (AMP)-activated protein kinase (AMPK), which is the central energy-sensing system of the cell. We established the fact that AMPK is activated upon glycolysis block resulting in mammalian target of rapamycin (mTOR) inhibition leading to Mcl-1 decrease, but no other Bcl-2 anti-apoptotic members. Interestingly, we determined that, upon glycolysis inhibition, the AMPK-mTOR pathway controlled Mcl-1 levels neither through transcriptional nor through posttranslational mechanism but rather by controlling its translation. Therefore, our results show a novel mechanism for the sensitization to DR-induced apoptosis linking glucose metabolism to Mcl-1 downexpression. In addition, this study provides a rationale for the combined use of DR ligands with AMPK activators or mTOR inhibitors in the treatment of human cancers.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis/physiology , Glycolysis/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Death Domain/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Antibodies/immunology , Antibodies/pharmacology , Apoptosis/drug effects , Blotting, Western , Deoxyglucose/pharmacology , Enzyme Activation/drug effects , Glucose/pharmacology , Glycolysis/drug effects , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Models, Biological , Myeloid Cell Leukemia Sequence 1 Protein , Protein Biosynthesis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA Interference , Receptors, Death Domain/immunology , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleotides/pharmacology , Sirolimus/pharmacology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , TOR Serine-Threonine Kinases , U937 Cells , fas Receptor/immunology , fas Receptor/metabolism
14.
Curr Opin Immunol ; 21(6): 708-14, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19837574

ABSTRACT

Downregulation of mast cells (MCs) function and/or survival is warranted in allergic inflammation (AI), mastocytosis/MC leukemias and in other inflammatory diseases in which MCs have a central role. Human MCs (hMCs) have been recently shown to express the death receptor (DR) TRAIL and the inhibitory receptors (IRs) CD300a and Siglec-8. TRAIL is the only known DR functional on hMCs, and interestingly its function is upregulated by IgE-dependent MC activation. The newly described IRs, CD300a and Siglec-8, potently downregulate MC activation and survival in vitro and inhibit different IgE-mediated responses in vivo. Therefore a selective targeting of TRAIL and of IRs on MC could be a novel immunopharmacological way to downregulate MC-associated diseases.


Subject(s)
Down-Regulation , Mast Cells/immunology , Animals , Antigens, CD/immunology , Humans , Receptors, Death Domain/immunology , TNF-Related Apoptosis-Inducing Ligand
15.
Nat Immunol ; 10(4): 348-55, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19295631

ABSTRACT

Death receptors (DRs) are members of the tumor necrosis factor receptor superfamily that possess a cytoplasmic death domain (DD). DRs regulate important operational and homeostatic aspects of the immune system. They transmit signals through apical protein complexes, which are nucleated by the DD adaptors FADD and TRADD, to control cellular outcomes that range from apoptosis to gene activation. FADD and TRADD also nucleate several distal signaling complexes, which mediate cross-talk between distinct DR signaling pathways. Moreover, together with other DR signal transducers, FADD and TRADD participate in functional complexes assembled by certain non-DR immune cell receptors, such as pattern-recognition receptors. Thus, DR signal transducers may provide important nodes of coordination in immune signaling networks.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Fas-Associated Death Domain Protein/physiology , Receptors, Death Domain/physiology , Signal Transduction/physiology , TNF Receptor-Associated Death Domain Protein/physiology , Adaptor Proteins, Signal Transducing/immunology , Animals , Apoptosis/physiology , Fas-Associated Death Domain Protein/immunology , Humans , Immunity, Active , Immunity, Innate , Receptors, Death Domain/immunology , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/physiology , TNF Receptor-Associated Death Domain Protein/immunology
16.
Immunopharmacol Immunotoxicol ; 31(2): 159-73, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18785049

ABSTRACT

Caspases are cysteine aspartases acting either as initiators (caspases 8, 9, and 10) or executioners (caspases 3, 6, and 7) to induce programmed cell death by apoptosis. Parasite infections by certain intracellular protozoans increase host cell life span by targeting caspase activation. Conversely, caspase activation, followed by apoptosis of lymphocytes and other cells, prevents effective immune responses to chronic parasite infection. Here we discuss how pharmacological inhibition of caspases might affect the immunity to protozoan infections, by either blocking or delaying apoptosis.


Subject(s)
Antiprotozoal Agents/therapeutic use , Apoptosis/drug effects , Caspase Inhibitors , Protozoan Infections/drug therapy , Animals , Antiprotozoal Agents/immunology , Apoptosis/immunology , Humans , Immune Tolerance/drug effects , Mice , Protozoan Infections/enzymology , Protozoan Infections/immunology , Receptors, Death Domain/immunology
17.
Autophagy ; 4(5): 715-6, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18560272

ABSTRACT

Activation of membrane death receptors has been connected to apoptosis and, recently, other non-apoptotic events. For example, we reported recently that sera from either a subset of patients with type 2 diabetes with neuropathy or a subpopulation of patients with neurogenic chronic intestinal pseudo-obstruction (CIP) stimulate autophagy in SH-SY5Y human neuroblastoma cells via complement-independent, autoantibody-mediated activation of Fas (CD95). Activation of the Fas pathway causes minimal activation of apoptosis in these cells since procaspase-8 shows low constitutive levels of expression in neuroblastoma cells. The observation that anti-Fas autoantibodies induce autophagy is novel and provocative. This finding has implications regarding the pathophysiology of diabetic neuropathy, CIP and, perhaps, other autoimmune disorders. For example, recent reports suggest that expression or activity of proapoptotic caspases can be enhanced by activation of more than one membrane death receptor, as could happen by combinations of cytokines and autoantibodies. The observation that autophagy, a putative cytoprotective pathway that has also been implicated in non-apoptotic cell death, is activated by autoantibodies against Fas, may represent an early cellular protective response. An increase in cytotoxic cytokine levels or the ratio of agonist:antagonist autoantibodies may "tip" the balance of the cellular response to activation of programmed cell death pathways.


Subject(s)
Autoantibodies/physiology , Autophagy/immunology , Receptors, Death Domain/physiology , Signal Transduction/immunology , Animals , Cell Line, Tumor , Diabetic Nephropathies/immunology , Humans , Receptors, Death Domain/immunology
18.
Oncogene ; 26(25): 3745-57, 2007 May 28.
Article in English | MEDLINE | ID: mdl-17530027

ABSTRACT

Deregulated cell death pathways may lead to the development of cancer, and induction of tumor cell apoptosis is the basis of many cancer therapies. Knowledge accumulated concerning the molecular mechanisms of apoptotic cell death has aided the development of new therapeutic strategies to treat cancer. Signals through death receptors of the tumor necrosis factor (TNF) superfamily have been well elucidated, and death receptors are now one of the most attractive therapeutic targets in cancer. In particular, DR5 and DR4, death receptors of TNF-related apoptosis-inducing ligand (TRAIL or Apo2L), are interesting targets of antibody-based therapy, since TRAIL may also bind decoy receptors that may prevent TRAIL-mediated apoptosis, whereas TRAIL ligand itself selectively induces apoptosis in cancer cells. Here, we review the potential therapeutic utility of agonistic antibodies against DR5 and DR4 and discuss the possible extension of this single-antibody-based strategy when combined with additional modalities that either synergizes to cause enhanced apoptosis or further engage the cellular immune response. Rational design of antibody-based therapies combining the induction of tumor cell apoptosis and activation of tumor-specific adaptive immunity enables promotion of distinct steps of the antitumor immune response, thereby enhancing tumor-specific lymphocytes that can eradicate TRAIL/DR5-resistant mutating, large established and heterogeneous tumors in a manner that does not require the definition of individual tumor-specific antigens.


Subject(s)
Neoplasms/drug therapy , Neoplasms/metabolism , Receptors, Death Domain/metabolism , Animals , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/therapeutic use , Disease Progression , Humans , Immunotherapy , Neoplasms/immunology , Neoplasms/pathology , Receptors, Death Domain/immunology , TNF-Related Apoptosis-Inducing Ligand/metabolism
19.
Mol Immunol ; 44(6): 1446-53, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16806475

ABSTRACT

The mechanisms responsible for the down-modulation of the activation of separated CD4(+) or CD8(+) human T cell blasts were studied using cells obtained from healthy donors. In the absence of IL-2, human CD4(+) T cell blasts were sensitive to both FasL and Apo2L/TRAIL, but human CD8(+) T cell blasts died, with no additional effect of death receptor ligation. CD8(+) T cell blasts were more sensitive than CD4(+) T cell blasts to apoptosis induction by IL-2 deprivation, which was associated with a decrease in the expression of anti-apoptotic proteins of the Bcl-2 family, especially of Mcl-1 in CD8(+) T cell blasts. The maintenance of high levels of Bim expression was also necessary, since down-modulation of Bim expression by siRNA in normal human CD8(+) T cell blasts greatly reduced apoptosis by IL-2 deprivation. These data, together with previous works, point to an important role of the presence or absence of immuno-stimulatory cytokines in the type of regulation of human CD8(+) T cell responses (death by cytokine deprivation versus death receptor inhibition of cytokine-dependent growth).


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Apoptosis/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Interleukin-2/deficiency , Lymphocyte Activation , Membrane Proteins/biosynthesis , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/biosynthesis , Receptors, Death Domain/metabolism , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Humans , Interleukin-2/genetics , Ligands , Membrane Proteins/genetics , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/immunology , RNA, Small Interfering/metabolism , Receptors, Death Domain/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...