Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Food Chem Toxicol ; 120: 143-154, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29990575

ABSTRACT

Dioscin has been known for its anti-cancer activity; however, its detailed molecular mechanisms have not been studied so far. Herein, we evaluated the anti-cancer activity of dioscin for proliferation inhibition and apoptosis in HepG2 cancer cells. Initially, dioscin was purified and identified from Polygonatum sibiricum by HPLC, MS, and NMR analysis, respectively. Dioscin inhibited the cell multiplication at IC50 of 8.34 µM, altered the cell morphology, arrested the cell cycle in G2/M phase and led to considerable programmed cell death. Furthermore, it has efficiently promoted the mitochondrial pathway and death receptor pathway. The inhibition of Caspase-8 and Caspase-9 proteins in these pathways abolished the dioscin induced apoptosis significantly; while dioscin inhibited the PI3K/Akt/mTOR pathway. Moreover, dioscin exposure led to enhanced intracellular ROS generation and the mRNA expression of JNK gene which emphasized their involvement in the apoptosis process in HepG2 cells.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cell Cycle Proteins/drug effects , Cell Division/drug effects , Diosgenin/analogs & derivatives , G2 Phase/drug effects , Genes, cdc/drug effects , Anticarcinogenic Agents/isolation & purification , Apoptosis/drug effects , Cell Proliferation/drug effects , Diosgenin/isolation & purification , Diosgenin/pharmacology , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Mitochondria/drug effects , Polygonatum/chemistry , Reactive Oxygen Species/metabolism , Receptors, Death Domain/drug effects , Receptors, Death Domain/metabolism
2.
Biomaterials ; 158: 56-73, 2018 03.
Article in English | MEDLINE | ID: mdl-29304403

ABSTRACT

Chemoresistance remains a formidable hurdle against cancer therapy. Seeking for novel therapy strategies is an urgent need for those who no longer benefit from chemotherapy. Chemoresistance is usually associated with the dysfunction of intrinsic apoptosis. Targeting extrinsic apoptosis via TRAIL signaling and the death receptors could be a potential solution to treat chemoresistant cancer. A highly biocompatible nano system for codelivery of the TRAIL DNA and the death receptor sensitizer monensin was developed, in which low-molecular-weight PEI (LMW-PEI) was crosslinked by the sulfhydryl cyclodextrin via disulfide bonds, and then bound with DNA, thus forming the bioreducible polyplex cores. In addition, the cyclodextrin also functioned as a carrier for the hydrophobic monensin via host-guest inclusion. Poly-γ-glutamic acid (γ-PGA) was used to modify the polyplex core via charge interaction. The γ-PGA corona can specifically bind with the tumor-associated gamma-glutamyl transpeptidase (GGT) overexpressed on the tumor cells, and achieve tumor-targeting delivery. Moreover, the tumor-homing peptide RGD-modified γ-PGA was also prepared as the surface coating materials for further improving gene delivery efficiency. This gene delivery system was characterized by the dual ligand-targeting, dual stimuli-responsive features. The ligands of RGD and γ-PGA can target the tumor-associated receptors (i.e., integrin and GGT). The conformation of γ-PGA is pH-sensitive, and the tumor acidic micro environments could trigger the detachment of surface-coating γ-PGA. The disulfide crosslinking LMW-PEI is redox-sensitive, and its fast disassembling in the tumor cells could favor the efficient gene delivery. The anti-tumor efficacy was demonstrated both in vitro and in vivo. Moreover, MYC-mediated synthetic lethality could be an important mechanism for overcoming the drug resistance. An important finding of our studies is the demonstration of the in vivo treatment efficacy of TRAIL/monensin, thus providing a potential novel therapeutic strategy for overcoming drug-resistant cancer.


Subject(s)
Drug Delivery Systems , Drug Resistance, Neoplasm , Monensin , Nanocomposites , Nanoparticles , TNF-Related Apoptosis-Inducing Ligand , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Humans , Imines/chemistry , Molecular Targeted Therapy , Monensin/administration & dosage , Nanocomposites/chemistry , Nanoparticles/chemistry , Polyethylenes/chemistry , Receptors, Death Domain/drug effects , Receptors, Death Domain/metabolism , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/genetics
3.
Drug Deliv ; 24(1): 1526-1536, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28994313

ABSTRACT

Active targeting nanoparticles were developed to simultaneously codeliver tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and Curcumin (Cur). In the nanoparticles (TRAIL-Cur-NPs), TRAIL was used as both active targeting ligand and therapeutic agent, and Cur could upregulate death receptors (DR4 and DR5) to increase the apoptosis-inducing effects of TRAIL. Compared with corresponding free drugs, TRAIL-Cur-NPs group showed enhanced cellular uptake, cytotoxicity and apoptosis induction effect on HCT116 colon cancer cells. In addition, in vivo anticancer studies suggested that TRAIL-Cur-NPs had superior therapeutic effect on tumors without obvious toxicity, which was mainly due to the high tumor targeting and synergistic effect of TRAIL and Cur. The synergistic mechanism of improved antitumor efficacy was proved to be upregulation of DR4 and DR5 in tumor cells induced by Cur. Thus, the prepared codelivery nanoparticles may have potential applications in colorectal cancer therapy.


Subject(s)
Apoptosis/drug effects , Nanoparticles/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Curcumin , Drug Liberation , Drug Synergism , Gene Expression , HCT116 Cells , Humans , Particle Size , Receptors, Death Domain/drug effects , Receptors, Death Domain/genetics , Surface Properties , TNF-Related Apoptosis-Inducing Ligand/pharmacokinetics , Up-Regulation
4.
Oncotarget ; 8(5): 8239-8249, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28030812

ABSTRACT

The fungal metabolites produced by Aspergillus flavus and Aspergillus parasiticus cause detrimental health effects on humans and animals. Particularly aflatoxin B1 (AFB1) is the most studied and a well-known global carcinogen, producing hepatotoxic, genotoxic and immunotoxic effects in multiple species. AFB1 is shown to provoke liver dysfunctioning by causing hepatocytes apoptosis and disturbing cellular enzymatic activities. In liver, AFB1 causes apoptosis via extrinsic mechanism because of high expression of death receptor pathway. The detailed mechanism of AFB1 induced hepatocytes apoptosis, via death receptor pathway still remains elusive. So the present study was conducted to explore apoptotic mechanism initiated by death receptors and associated genes in aflatoxin B1 induced liver apoptosis in chickens fed with AFB1 for 3 weeks. Results from the present study displayed histopathological and ultrastructural changes in liver such as hydropic degeneration, fatty vacuolar degeneration and proliferation of bile duct in hepatocytes in AFB1 group, along with imbalance between reactive oxygen species (ROS) and antioxidant defense system upon AFB1 ingestion. Moreover, AFB1 intoxicated chickens showed upregulation of death receptors FAS, TNFR1 and associated genes and downregulation of inhibitory apoptotic proteins XIAP and BCL-2. The results obtained from this novel and comprehensive study including histopathological, ultrastructural, flow cytometrical and death receptor pathway gene expression profiles, will facilitate better understanding of mechanisms and involvement of death receptor pathway in hepatocytes apoptosis induced by AFB1 and ultimately may be helpful in bringing down the toxigenic potential of AFB1.


Subject(s)
Aflatoxin B1/toxicity , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/etiology , Hepatocytes/drug effects , Liver/drug effects , Receptors, Death Domain/drug effects , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chickens , Gene Expression Regulation , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Liver/metabolism , Liver/ultrastructure , Receptors, Death Domain/genetics , Receptors, Death Domain/metabolism , Signal Transduction/drug effects , Time Factors
5.
Sci Rep ; 5: 18629, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26686000

ABSTRACT

Bio-functionalized nanoparticles with semiconducting/metallic core encapsulated in a bio- or bio-derived materials are promising for applications in biology and especially in cancer diagnostic and healing. In this report, we report a facile, single-step, first-time synthesis and in-situ functionalization strategy for the preparation of monodispersed selenium nanoparticles (SeNPs) functionalized using a novel polysaccharide (DP1) extracted from Dictyophora indusiata (a fungus). The DP1 functionalized SeNPs (DP1-SeNPs), where DP1 is attached to the surface via Se-O bond as well as physic-sorption had, an average diameter of 89 nm, and were highly uniform, extremely stable compared to bare SeNPs. Detailed investigation of the biological properties of DP1-SeNP illustrated that they exhibit unprecedented, enhanced, and selective antiproliferative activity through inducing cell apoptosis confirmed by nuclear condensation, DNA cleavage, and accumulation of S phase cell arrest. The mechanism of the induced apoptosis was found to be a combination of the activation of caspases 3, 8, and 9, the Fas-associated death domain protein (FADD), reactive oxygen species (ROS) overproduction, as well as mitochondrial dysfunction. It is envisioned that the reported DP1-SeNPs will offer a new phase space for high-efficiency anticancer treatment with little side effect.


Subject(s)
Cell Proliferation/drug effects , Nanoparticles/administration & dosage , Polysaccharides/pharmacology , Selenium/pharmacology , Apoptosis/drug effects , Basidiomycota/chemistry , Drug Delivery Systems , Hep G2 Cells , Humans , Mitochondria/drug effects , Mitochondria/pathology , Nanoparticles/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Polysaccharides/chemistry , Reactive Oxygen Species/metabolism , Receptors, Death Domain/drug effects , S Phase/drug effects , Selenium/chemistry , Signal Transduction/drug effects
6.
Eur J Pharmacol ; 761: 309-20, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26086857

ABSTRACT

Nortriptyline (NTP), an antidepressant, has antitumor effects on some human cancer cells, but its effect on human bladder cancer cells is not known. In this study, we used a cell viability assay to demonstrate that NTP is cytotoxic to human TCCSUP and mouse MBT-2 bladder cancer cells in a concentration and time-dependent manner. We also performed cell cycle analysis, annexin V and mitochondrial membrane potential assays, and Western blot analysis to show that NTP inhibits cell growth in these cells by inducing both mitochondria-mediated and death receptor-mediated apoptosis. Specifically, NTP increases the expression of Fas, FasL, FADD, Bax, Bak, and cleaved forms of caspase-3, caspase-8, caspase-9, and poly(ADP-ribose) polymerase. In addition, NTP decreases the expression of Bcl-2, Bcl-xL, BH3 interacting domain death agonist, X-linked inhibitor of apoptosis protein, and survivin. Furthermore, NTP-induced apoptosis is associated with reactive oxygen species (ROS) production, which can be reduced by antioxidants, such as N-acetyl-L-cysteine. Finally, we showed that NTP suppresses tumor growth in mice inoculated with MBT-2 cells. Collectively, our results suggest that NTP induces both intrinsic and extrinsic apoptosis in human and mouse bladder cancer cells and that it may be a clinically useful chemotherapeutic agent for bladder cancer in humans.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Mitochondria/drug effects , Nortriptyline/pharmacology , Receptors, Death Domain/drug effects , Tumor Burden/drug effects , Urinary Bladder Neoplasms/drug therapy , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Male , Mice, Inbred C3H , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Receptors, Death Domain/metabolism , Signal Transduction/drug effects , Time Factors , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
7.
Reproduction ; 150(2): 105-14, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25934945

ABSTRACT

Orchitis (testicular swelling) often occurs during systemic inflammatory conditions, such as sepsis. Interleukin 18 (IL18) is a proinflammatory cytokine and is an apoptotic mediator during endotoxemia, but the role of IL18 in response to inflammation in the testes was unclear. WT and IL18 knockout (KO) mice were injected lipopolysaccharide (LPS) to induce endotoxemia and examined 12 and 48  h after LPS administration to model the acute and recovery phases of endotoxemia. Caspase activation was assessed using immunohistochemistry. Protein and mRNA expression were examined by western blot and quantitative real-time RT-PCR respectively. During the acute phase of endotoxemia, apoptosis (as indicated by caspase-3 cleavage) was increased in WT mice but not in IL18 KO mice. The death receptor-mediated and mitochondrial-mediated apoptotic pathways were both activated in the WT mice but not in the KO mice. During the recovery phase of endotoxemia, apoptosis was observed in the IL18 KO mice but not in the WT mice. Activation of the death-receptor mediated apoptotic pathway could be seen in the IL18 KO mice but not the WT mice. These results suggested that endogenous IL18 induces germ cell apoptosis via death receptor mediated- and mitochondrial-mediated pathways during the acute phase of endotoxemia and suppresses germ cell apoptosis via death-receptor mediated pathways during recovery from endotoxemia. Taken together, IL18 could be a new therapeutic target to prevent orchitis during endotoxemia.


Subject(s)
Apoptosis/drug effects , Endotoxemia/pathology , Germ Cells/drug effects , Interleukin-18/pharmacology , Testis/drug effects , Animals , Behavior, Animal/drug effects , Caspases/metabolism , Enzyme Activation/drug effects , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orchitis/chemically induced , Orchitis/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Death Domain/drug effects , Receptors, Death Domain/genetics , Shock, Septic/chemically induced , Shock, Septic/psychology , Testis/cytology
8.
Food Chem Toxicol ; 71: 51-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24930757

ABSTRACT

TRAIL induces apoptosis in a variety of tumor cells. However, development of resistance to TRAIL is a major obstacle to more effective cancer treatment. Therefore, novel pharmacological agents that enhance sensitivity to TRAIL are necessary. In the present study, we investigated the molecular mechanisms by which ilimaquinone isolated from a sea sponge sensitizes human colon cancer cells to TRAIL. Ilimaquinone pretreatment significantly enhanced TRAIL-induced apoptosis in HCT 116 cells and sensitized colon cancer cells to TRAIL-induced apoptosis through increased caspase-8, -3 activation, PARP cleavage, and DNA damage. Ilimaquinone also reduced the cell survival proteins Bcl2 and Bcl-xL, while strongly up-regulating death receptor (DR) 4 and DR5 expression. Induction of DR4 and DR5 by ilimaquinone was mediated through up-regulation of CCAAT/enhancer-binding protein homologous protein (CHOP). The up-regulation of CHOP, DR4 and DR5 expression was mediated through activation of extracellular-signal regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) signaling pathways. Finally, the generation of ROS was required for CHOP and DR5 up-regulation by ilimaquinone. These results demonstrate that ilimaquinone enhanced the sensitivity of human colon cancer cells to TRAIL-induced apoptosis through ROS-ERK/p38 MAPK-CHOP-mediated up-regulation of DR4 and DR5 expression, suggesting that ilimaquinone could be developed into an adjuvant chemotherapeutic drug.


Subject(s)
Apoptosis/physiology , Colonic Neoplasms/pathology , Quinones/toxicity , Receptors, Death Domain/drug effects , Sesquiterpenes/toxicity , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/physiology , Base Sequence , Cell Line, Tumor , DNA Primers , Enzyme Activation , Humans , Mitogen-Activated Protein Kinases/metabolism , Proton Magnetic Resonance Spectroscopy , Quinones/chemistry , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Death Domain/metabolism , Sesquiterpenes/chemistry , Transcription Factor CHOP/metabolism , Up-Regulation/drug effects
9.
Antimicrob Agents Chemother ; 58(7): 4075-85, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24798292

ABSTRACT

Nephrotoxicity is the dose-limiting factor for colistin, but the exact mechanism is unknown. This study aimed to investigate the roles of the mitochondrial, death receptor, and endoplasmic reticulum pathways in colistin-induced nephrotoxicity. Mice were intravenously administered 7.5 or 15 mg of colistin/kg of body weight/day (via a 3-min infusion and divided into two doses) for 7 days. Renal function, oxidative stress, and apoptosis were measured. Representative biomarkers involved in the mitochondrial, death receptor, and endoplasmic reticulum pathways were investigated, and the key markers involved in apoptosis and autophagy were examined. After 7-day colistin treatment, significant increase was observed with blood urea nitrogen, serum creatinine, and malondialdehyde, while activities of superoxide dismutase (SOD) and catalase decreased in the kidneys. Acute tubular necrosis and mitochondrial dysfunction were detected, and colistin-induced apoptosis was characterized by DNA fragmentation, cleavage of poly(ADP-ribose) polymerase (PARP-1), increase of 8-hydroxydeoxyguanosine (8-OHdG), and activation of caspases (caspase-8, -9, and -3). It was evident that colistin-induced apoptosis involved the mitochondrial pathway (downregulation of Bcl-2 and upregulation of cytochrome C [cytC] and Bax), death receptor pathway (upregulation of Fas, FasL, and Fas-associated death domain [FADD]), and endoplasmic reticulum pathway (upregulation of Grp78/Bip, ATF6, GADD153/CHOP, and caspase-12). In the 15-mg/kg/day colistin group, expression of the cyclin-dependent kinase 2 (CDK2) and phosphorylated JNK (p-JNK) significantly increased (P < 0.05), while in the 7.5-mg/kg/day colistin group, a large number of autophagolysosomes and classic autophagy were observed. Western blot results of Beclin-1 and LC3B indicated that autophagy may play a protective role in colistin-induced nephrotoxicity. In conclusion, this is the first study to demonstrate that all three major apoptosis pathways and autophagy are involved in colistin-induced nephrotoxicity.


Subject(s)
Colistin/toxicity , Endoplasmic Reticulum/metabolism , Kidney/pathology , Mitochondria/metabolism , Receptors, Death Domain/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Beclin-1 , Biomarkers/metabolism , Blood Urea Nitrogen , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Catalase/metabolism , Creatinine/blood , DNA Fragmentation/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation/drug effects , Female , Malondialdehyde/blood , Mice , Microtubule-Associated Proteins/biosynthesis , Mitochondria/drug effects , Oxidative Stress/drug effects , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Receptors, Death Domain/drug effects , Superoxide Dismutase/metabolism
10.
Expert Rev Anticancer Ther ; 14(6): 641-4, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24791860

ABSTRACT

Metastasis contributes to over 90% of cancer-related deaths. Many types of cancer metastasize via the bloodstream, where circulating tumor cells (CTCs) originating from the primary tumor can undergo selectin-mediated adhesion with the blood vessel wall and subsequently transmigrate to anatomically distant organs. In an effort to neutralize CTCs with the potential to form metastases, a new therapeutic approach has been developed in which circulating leukocytes are functionalized to target and kill cancer cells in the bloodstream. This approach mimics the cytotoxic activity of natural killer cells and the chemical engineering concept of a fluidized bed reactor, which increases the surface area for surface-catalyzed reactions. The resulting 'unnatural killer cells', proven effective in vitro with human blood and also in the living mouse, holds promise in neutralizing CTCs to interrupt the metastasis process.


Subject(s)
E-Selectin/administration & dosage , Leukocytes/immunology , Neoplasm Metastasis/prevention & control , Neoplastic Cells, Circulating , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Animals , Apoptosis/drug effects , Cell Adhesion , Cytotoxicity, Immunologic , E-Selectin/metabolism , Erythrocytes/physiology , Hemorheology , Humans , Leukocytes/drug effects , Liposomes , Mice , Nanostructures , Neoplasm Metastasis/physiopathology , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Receptors, Death Domain/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology
11.
Br J Pharmacol ; 169(8): 1723-44, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23638798

ABSTRACT

UNLABELLED: Anti-tumour therapies based on the use pro-apoptotic receptor agonists, including TNF-related apoptosis-inducing ligand (TRAIL) or monoclonal antibodies targeting TRAIL-R1 or TRAIL-R2, have been disappointing so far, despite clear evidence of clinical activity and lack of adverse events for the vast majority of these compounds, whether combined or not with conventional or targeted anti-cancer therapies. This brief review aims at discussing the possible reasons for the lack of apparent success of these therapeutic approaches and at providing hints in order to rationally design optimal protocols based on our current understanding of TRAIL signalling regulation or resistance for future clinical trials. LINKED ARTICLES: This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Receptors, Death Domain/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/drug effects , Death Domain Receptor Signaling Adaptor Proteins/drug effects , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Humans , Recombinant Proteins , TNF-Related Apoptosis-Inducing Ligand/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/therapeutic use
12.
Cell Death Differ ; 20(7): 858-68, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23579241

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-based therapy is currently evaluated in clinical studies as a tumor cell selective pro-apoptotic approach. However, besides activating canonical caspase-dependent apoptosis by binding to TRAIL-specific death receptors, the TRAIL ligand can activate non-canonical cell survival or proliferation pathways in resistant tumor cells through the same death receptors, which is counterproductive for therapy. Even more, recent studies indicate metastases-promoting activity of TRAIL. In this review, the remarkable dichotomy in TRAIL signaling is highlighted. An overview of the currently known mechanisms involved in non-canonical TRAIL signaling and the subsequent activation of various kinases is provided. These kinases include RIP1, IκB/ NF-κB, MAPK p38, JNK, ERK1/2, MAP3K TAK1, PKC, PI3K/Akt and Src. The functional consequences of their activation, often being stimulation of tumor cell survival and in some cases enhancement of their invasive behavior, are discussed. Interestingly, the non-canonical responses triggered by TRAIL in resistant tumor cells resemble that of TRAIL-induced signals in non-transformed cells. Better knowledge of the mechanism underlying the dichotomy in TRAIL receptor signaling may provide markers for selecting patients who will likely benefit from TRAIL-based therapy and could provide a rationalized basis for combination therapies with TRAIL death receptor-targeting drugs.


Subject(s)
Neoplasms/pathology , Neoplasms/physiopathology , Phosphotransferases/physiology , Receptors, Death Domain/physiology , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Disease Models, Animal , Humans , I-kappa B Kinase/physiology , Mice , Mitogen-Activated Protein Kinase Kinases/physiology , NF-kappa B/physiology , Neoplasms/drug therapy , Receptors, Death Domain/drug effects , TNF-Related Apoptosis-Inducing Ligand/drug effects
13.
Neurotoxicology ; 33(3): 416-23, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22531227

ABSTRACT

We compared the neurotoxic effects of 14 nm silver nanoparticles (AgNPs) and ionic silver, in the form of silver acetate (AgAc), in vivo and in vitro. In female rats, we found that AgNPs (4.5 and 9 mg AgNP/kg bw/day) and ionic silver (9 mg Ag/kg bw/day) increased the dopamine concentration in the brain following 28 days of oral administration. The concentration of 5-hydroxytryptamine (5-HT) in the brain was increased only by AgNP at a dose of 9 mg Ag/kg bw/day. Only AgAc (9 mg Ag/kg bw/day) was found to increase noradrenaline concentration in the brain. In contrast to the results obtained from a 28-day exposure, the dopamine concentration in the brain was decreased by AgNPs (2.25 and 4.5mg/kg bw/day) following a 14-day exposure. These data suggest that there are differential effects of silver on dopamine depending on the length of exposure. In vitro, AgNPs, AgAc and a 12 kDa filtered sub-nano AgNP fraction were used to investigate cell death mechanisms in neuronal-like PC12 cells. AgNPs and the 12 kDa filtered fraction decreased cell viability to a similar extent, whereas AgAc was relatively more potent. AgNPs did not induce necrosis. However, apoptosis was found to be equally increased in cells exposed to AgNPs and the 12kDa filtered fraction, with AgAc showing a greater potency. Both the mitochondrial and the death receptor pathways were found to be involved in AgNP- and AgAc-induced apoptosis. In conclusion, 14 nm AgNPs and AgAc affected brain neurotransmitter concentrations. AgNP affected 5-HT, AgAc affected noradrenaline, whereas both silver formulations affected dopamine. Furthermore, apoptosis was observed in neuronal-like cells exposed to AgNPs, a 12 kDa filtered fraction of AgNP, and AgAc. These findings suggest that ionic silver and a 14 nm AgNP preparation have similar neurotoxic effects; a possible explanation for this could be the release and action of ionic silver from the surface of AgNPs.


Subject(s)
Acetates/toxicity , Brain/drug effects , Metal Nanoparticles , Neurons/drug effects , Silver Compounds/toxicity , Acetates/metabolism , Animals , Apoptosis/drug effects , Brain/metabolism , Brain/pathology , Cell Survival/drug effects , Dopamine/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Female , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Norepinephrine/metabolism , PC12 Cells , Particle Size , Rats , Rats, Wistar , Receptors, Death Domain/drug effects , Receptors, Death Domain/metabolism , Serotonin/metabolism , Silver Compounds/metabolism , Time Factors , Up-Regulation
14.
Invest New Drugs ; 30(2): 548-57, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21120579

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) induces apoptosis in various cancer cells. Diarylheptanoids such as hirsutenone and oregonin have been shown to have anti-inflammatory and anti-tumor effects. However, it is still unknown by which mechanism diarylheptanoids induce cell death. In addition, the effect of hirsutenone on TRAIL-induced apoptosis in the human epithelial ovarian carcinoma cell lines is unknown. To assess the apoptosis promoting effect of hirsutenone, we investigated the effect of hirsutenone on the apoptotic effect of TRAIL using the human epithelial carcinoma cell lines OVCAR-3 and SK-OV-3. TRAIL induced nuclear damage, decrease in Bid, Bcl-2 and Bcl-xL protein levels, increase in Bax levels, loss of the mitochondrial transmembrane potential, cytochrome c release, activation of caspases (8, 9 and 3) and increase in tumor suppressor p53 levels. Hirsutenone enhanced the TRAIL-induced apoptosis-related protein activation, nuclear damage and cell death. The results suggest that hirsutenone may enhance the apoptotic effect of TRAIL on ovarian carcinoma cell lines by increasing the activation of the caspase-8- and Bid-dependent pathways and the mitochondria-mediated apoptotic pathway, leading to caspase activation. Hirsutenone may confer a benefit in the TRAIL treatment of epithelial ovarian adenocarcinoma.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Catechols/pharmacology , Diarylheptanoids/pharmacology , Mitochondria/drug effects , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Receptors, Death Domain/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Receptors, Death Domain/metabolism , Recombinant Proteins/pharmacology
15.
Recent Pat Anticancer Drug Discov ; 7(2): 207-17, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22114983

ABSTRACT

Multicellular organisms require apoptosis whereby the human body eliminates unnecessary and/or damaged cells. Apoptosis, or programmed cell death, can indeed be considered as a constitutive anti-cancer mechanism that seems to be defective in more than 50% of cancers. Molecular insights on the biology of the apoptotic process have led to the development of new anti-cancer strategies aiming at recovering and stimulating this process. Preclinical and clinical studies of our and other groups have demonstrated that targeting the extrinsic apoptotic pathway by various death receptors agonists is a safe and effective anti-cancer strategy, which thus may become a new cornerstone of cancer therapy. Here, we review the most recent acquisitions and patents on TRAIL or TRAIL mimetics, as well as the combination therapies that could be used with them.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Neoplasms/drug therapy , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols , Clinical Trials as Topic , Humans , Mice , Receptors, Death Domain/drug effects , TNF-Related Apoptosis-Inducing Ligand/agonists , TNF-Related Apoptosis-Inducing Ligand/chemistry , TNF-Related Apoptosis-Inducing Ligand/metabolism
16.
Med Hypotheses ; 77(1): 144-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21507581

ABSTRACT

Retinal detachment (RD) is a common cause of human visual impairment. Detachment of photoreceptors from the retinal pigment epithelium causes photoreceptor loss and subsequent vision decline. Death receptor (DR)-induced apoptosis play critical role in activating apoptosis in photoreceptor cells. Z-VAD-FMK inhibits the DR-induced retinal neuronal apoptosis but promotes neuronal death through necroptosis pathway, an alternative programmed cell death, which can be inhibited by Nec-1. Thus, we may achieve a better result by simultaneous inhibition of DR-induced apoptosis and necroptosis, which provides us with a new direction in the treatment of RD.


Subject(s)
Apoptosis , Neurons/pathology , Receptors, Death Domain/drug effects , Retinal Detachment/pathology , Humans , Necrosis , Receptors, Death Domain/metabolism , Receptors, Death Domain/physiology , Signal Transduction
17.
J Nat Prod ; 74(4): 866-70, 2011 Apr 25.
Article in English | MEDLINE | ID: mdl-21381696

ABSTRACT

Thirty-five limonoids, including 15 of the azadiradione type (1-15), five of the gedunin type (16-20), four of the azadirachtin type (21-24), nine of the nimbin type (25-33), and two degraded limonoids (34, 35), isolated from Azadirachta indica seed extracts, were evaluated for their cytotoxic activities against five human cancer cell lines. Seven compounds (3, 6, 7, 16, 18, 28, and 29) exhibited cytotoxic activity against one or more cell lines. Among these compounds, 7-deacetyl-7-benzoylepoxyazadiradione (7), 7-deacetyl-7-benzoylgeduin (18), and 28-deoxonimbolide (28) exhibited potent cytotoxic activity against HL60 leukemia cells with IC(50) values in the range 2.7-3.1 µM. Compounds 7, 18, and 28 induced early apoptosis in HL60 cells, observed by flow cytometry. Western blot analysis showed that compounds 7, 18, and 28 activated caspases-3, -8, and -9 in HL60 cells. This suggested that compounds 7, 18, and 28 induced apoptotic cell death in HL60 cells via both the mitochondrial- and the death receptor-mediated pathways. Futhermore, compound 7 was shown to possess high selective cytotoxicity for leukemia cells since it exhibited only weak cytotoxicity against a normal lymphocyte cell line (RPMI 1788).


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Azadirachta/chemistry , Limonins/isolation & purification , Limonins/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Drug Screening Assays, Antitumor , HL-60 Cells , Humans , Limonins/chemistry , Lymphocytes/drug effects , Mitochondria/drug effects , Molecular Structure , Receptors, Death Domain/drug effects , Seeds/chemistry
18.
Anticancer Drugs ; 22(6): 481-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21317766

ABSTRACT

Death receptors play a crucial role in immune surveillance and cellular homeostasis, two processes circumvented by tumor cells. CD95 (also termed Fas or APO1) is a transmembrane receptor, which belongs to the tumor necrosis factor receptor superfamily, and induces a potent apoptotic signal. Initial steps of the CD95 signal take place through protein/protein interactions that bring zymogens such as caspase-8 and caspase-10 closer. Aggregation of these procaspases leads to their autoprocessing, to the release of activated caspases in the cytosol, which causes a caspase cascade, and to the transmission of the apoptotic signal. In parallel, CD95 engagement drives an increase in the intracellular calcium concentration (Ca(2+))i whose origin and functions remain controversial. Although Ca(2+) ions play a central role in apoptosis/necrosis induction, recent studies have highlighted a protective role of Ca(2+) in death receptor signaling. In the light of these findings, we discuss the role of Ca(2+) ions as modulators of CD95 signaling.


Subject(s)
Calcium/physiology , Signal Transduction/physiology , fas Receptor/physiology , Apoptosis/drug effects , Caspases/metabolism , Humans , Receptors, Death Domain/drug effects
19.
Eur Urol ; 59(5): 734-44, 2011 May.
Article in English | MEDLINE | ID: mdl-21269758

ABSTRACT

BACKGROUND: Quinazoline-based α1-adrenoceptor antagonists suppress tumor growth by inducing apoptosis via an α1-adrenoceptor-independent action. Anoikis is a unique mode of apoptosis consequential to insufficient cell-matrix interactions. OBJECTIVE: This study investigated the apoptotic effect of novel quinazoline-based compounds on human renal cancer cells. DESIGN, SETTING, AND PARTICIPANTS: Two cell lines were used: renal cell carcinoma (RCC) 786-0, harboring a von Hippel-Lindau (VHL) tumor-suppressor gene mutation with a highly angiogenic phenotype, and Caki cells (no VHL mutation). MEASUREMENTS: The lead compound DZ-50 (10 µM) led to significant inhibition of tumor-cell adhesion, migration, and invasion at a lower dose than doxazosin (25 µM) in both RCC lines. RESULTS AND LIMITATIONS: Doxazosin induced death-receptor-mediated apoptosis, while DZ-50 led to anoikis via targeting of the focal adhesion complex and AKT signaling that subsequently increased RCC susceptibility to caspase-8-mediated apoptosis. Both quinazoline compounds, doxazosin and DZ-50, significantly reduced RCC metastatic potential in vivo. CONCLUSIONS: Quinazoline-based drugs trigger anoikis in RCC by targeting the focal adhesion survival signaling. This potent antitumor action against human RCC suggests a novel quinazoline-based therapy targeting renal cancer.


Subject(s)
Anoikis/drug effects , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/enzymology , Doxazosin/analogs & derivatives , Focal Adhesion Kinase 1/metabolism , Kidney Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Caspase 8/metabolism , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Doxazosin/pharmacology , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mutation , Neoplasm Invasiveness , Receptors, Death Domain/drug effects , Receptors, Death Domain/metabolism , Time Factors , Transendothelial and Transepithelial Migration/drug effects , Von Hippel-Lindau Tumor Suppressor Protein/genetics
20.
Toxicol In Vitro ; 24(7): 1920-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20696234

ABSTRACT

The pro-apoptotic activity of J-7, a synthetic methyl jasmonate derivative, on the Hep3B human hepatocarcinoma cell line was investigated. Treatment of Hep3B cells with J-7 resulted in growth inhibition and the induction of apoptosis as measured by trypan blue-excluding cells, MTT assay, nuclear staining, DNA fragmentation, and flow cytometry analysis. The increased apoptotic events in Hep3B cells caused by J-7 were associated with the alteration in the ratio of Bax/Bcl-2 protein expression. J-7 treatment induced the expression of death receptor-related proteins such as death receptor 5, which triggered the activation of caspase-8 and the down-regulation of the whole Bid expression. In addition, the apoptosis induction by J-7 was correlated with the activation of caspase-9 and caspase-3, down-regulation IAP family proteins such as XIAP and cIAP-1, and concomitant degradation of poly (ADP-ribose) polymerase. However, the cytotoxic and apoptotic effects induced by J-7 were significantly inhibited by z-DEVD-fmk, a caspase-3 inhibitor, which demonstrates the important role that caspase-3 plays in the process. Furthermore, blocking the extracellular signal-regulated protein kinase and c-Jun N-terminal kinase pathways showed increased apoptosis and the activation of caspases in J-7-induced apoptosis. The results indicated that J-7 induces the apoptosis of Hep3B cells through a signaling cascade of death-receptor-mediated extrinsic as well as mitochondria-mediated intrinsic caspase pathways, which are associated with the activation of the mitogen-activated protein kinases signal pathway.


Subject(s)
Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Cyclopentanes/pharmacology , Fatty Acids, Unsaturated/pharmacology , Liver Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Caspases/drug effects , Caspases/metabolism , Cell Line, Tumor , DNA Fragmentation/drug effects , Flow Cytometry , Humans , Liver Neoplasms/pathology , Mitochondria/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Receptors, Death Domain/drug effects , Receptors, Death Domain/genetics , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...