301
|
|
302
|
Affiliation(s)
- Nima Rezaei
- Children's Medical Center Hospital, Tehran University of Medical Sciences Research Center for Immunodeficiencies, Tehran, Iran
| |
Collapse
|
303
|
Ahmed SMU, Wu X, Jin X, Zhang X, Togo Y, Suzuki T, Li Y, Kanematsu A, Nojima M, Yamamoto S, Sugimoto M, Kakehi Y. Synergistic induction of apoptosis by mapatumumab and anthracyclines in human bladder cancer cells. Oncol Rep 2014; 33:566-72. [PMID: 25483927 DOI: 10.3892/or.2014.3654] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 11/21/2014] [Indexed: 12/16/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) triggers apoptosis in a variety of tumor cells by engaging the death receptors 4 (DR4) and 5 (DR5). We investigated the effect of chemotherapeutic drugs on DR4-mediated apoptosis in human bladder cancer cells, using a human monoclonal agonistic antibody specific for DR4, mapatumumab. Cytotoxicity was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Synergy was assessed by isobolographic analysis. Treatment of human bladder cancer T24 cells with mapatumumab in combination with mitomycin C, vinblastine or gemcitabine did not overcome resistance to these agents. However, treatment with mapatumumab in combination with epirubicin (EPI) had a synergistic cytotoxic effect. Synergy was also obtained in KU7 and RT112 human bladder cancer cells. A synergistic effect was also observed with mapatumumab in combination with pirarubicin. The synergy obtained in cytotoxicity with mapatumumab and EPI was also achieved in apoptosis. EPI markedly increased DR4 expression in the bladder cancer cells at both the mRNA and protein levels. Furthermore, the combination-induced cytotoxicity was significantly suppressed by the DR4:Fc chimeric protein. The combination of EPI and mapatumumab significantly activated the caspase cascade, including caspase-8, -9 and -3, which are the downstream molecules of death receptors. These findings indicate that EPI sensitizes bladder cancer cells to DR4-mediated apoptosis through induction of DR4 and activation of caspases, suggesting that the combination therapy of EPI and mapatumumab may be effective for bladder cancer therapy.
Collapse
Affiliation(s)
- Syed Minhaj Uddin Ahmed
- Department of Urology, Faculty of Medicine, Kagawa University, Kita-gun, Kagawa 761-0793, Japan
| | - Xiuxian Wu
- Department of Urology, Hyogo Medical College, Nishinomiya City, Hyogo 663-8501, Japan
| | - Xinghua Jin
- Department of Urology, Hyogo Medical College, Nishinomiya City, Hyogo 663-8501, Japan
| | - Xia Zhang
- Department of Urology, Faculty of Medicine, Kagawa University, Kita-gun, Kagawa 761-0793, Japan
| | - Yoshikazu Togo
- Department of Urology, Hyogo Medical College, Nishinomiya City, Hyogo 663-8501, Japan
| | - Toru Suzuki
- Department of Urology, Hyogo Medical College, Nishinomiya City, Hyogo 663-8501, Japan
| | - Yongnan Li
- Department of Urology, Hyogo Medical College, Nishinomiya City, Hyogo 663-8501, Japan
| | - Akihiro Kanematsu
- Department of Urology, Hyogo Medical College, Nishinomiya City, Hyogo 663-8501, Japan
| | - Mikio Nojima
- Department of Urology, Hyogo Medical College, Nishinomiya City, Hyogo 663-8501, Japan
| | - Shingo Yamamoto
- Department of Urology, Hyogo Medical College, Nishinomiya City, Hyogo 663-8501, Japan
| | - Mikio Sugimoto
- Department of Urology, Faculty of Medicine, Kagawa University, Kita-gun, Kagawa 761-0793, Japan
| | - Yoshiyuki Kakehi
- Department of Urology, Faculty of Medicine, Kagawa University, Kita-gun, Kagawa 761-0793, Japan
| |
Collapse
|
304
|
Lawrie A. The role of the osteoprotegerin/tumor necrosis factor related apoptosis-inducing ligand axis in the pathogenesis of pulmonary arterial hypertension. Vascul Pharmacol 2014; 63:114-7. [PMID: 25446166 DOI: 10.1016/j.vph.2014.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 09/25/2014] [Accepted: 10/04/2014] [Indexed: 12/14/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal condition driven by a progressive remodelling of the small pulmonary arteries through sustained vasoconstriction, and vascular cell proliferation. This process causes a substantial reduction in luminal area increasing pulmonary vascular resistance and blood pressure leading to right heart failure. Current medical therapies can alleviate some symptoms and reduce the vasoconstrictive aspects of disease but new treatments are required that target the vascular cell proliferation if we are to develop new therapies. Expression of the tumour necrosis factor related apoptosis-inducing ligand (TRAIL) and osteoprotegerin (OPG) proteins are increased in IPAH. Specifically OPG is increased within the serum of patients with idiopathic pulmonary arterial hypertension (IPAH) and has prognostic utility, and both OPG and TRAIL are increased within pulmonary vascular lesions of patients with IPAH, and are mitogens for pulmonary artery smooth muscle cells in vitro. We have demonstrated that genetic deletion, or antibody blockade of TRAIL prevents, and critically reverses the development of PAH in multiple rodent models. The role OPG plays in this process both through interacting with TRAIL, and indirectly through other mechanisms is currently unclear these but data highlight the critical importance of this pathway in PAH pathogenesis, and its potential for future therapies.
Collapse
Affiliation(s)
- Allan Lawrie
- Department of Cardiovascular Science, Faculty of Medicine, Dentistry & Health, University of Sheffield, Sheffield S10 2RX, United Kingdom.
| |
Collapse
|
305
|
Sarhan D, D'Arcy P, Lundqvist A. Regulation of TRAIL-receptor expression by the ubiquitin-proteasome system. Int J Mol Sci 2014; 15:18557-73. [PMID: 25318057 PMCID: PMC4227232 DOI: 10.3390/ijms151018557] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 09/30/2014] [Accepted: 10/02/2014] [Indexed: 12/21/2022] Open
Abstract
The tumor necrosis factor (TNF)-related apoptosis-inducing ligand- receptor (TRAIL-R) family has emerged as a key mediator of cell fate and survival. Ligation of TRAIL ligand to TRAIL-R1 or TRAIL-R2 initiates the extrinsic apoptotic pathway characterized by the recruitment of death domains, assembly of the death-inducing signaling complex (DISC), caspase activation and ultimately apoptosis. Conversely the decoy receptors TRAIL-R3 and TRAIL-R4, which lack the pro-apoptotic death domain, function to dampen the apoptotic response by competing for TRAIL ligand. The tissue restricted expression of the decoy receptors on normal but not cancer cells provides a therapeutic rational for the development of selective TRAIL-mediated anti-tumor therapies. Recent clinical trials using agonistic antibodies against the apoptosis-inducing TRAIL receptors or recombinant TRAIL have been promising; however the number of patients in complete remission remains stubbornly low. The mechanisms of TRAIL resistance are relatively unexplored but may in part be due to TRAIL-R down-regulation or shedding of TRAIL-R by tumor cells. Therefore a better understanding of the mechanisms underlying TRAIL resistance is required. The ubiquitin-proteasome system (UPS) has been shown to regulate TRAIL-R members suggesting that pharmacological inhibition of the UPS may be a novel strategy to augment TRAIL-based therapies and increase efficacies. We recently identified b-AP15 as an inhibitor of proteasome deubiquitinase (DUB) activity. Interestingly, exposure of tumor cell lines to b-AP15 resulted in increased TRAIL-R2 expression and enhanced sensitivity to TRAIL-mediated apoptosis and cell death in vitro and in vivo. In conclusion, targeting the UPS may represent a novel strategy to increase the cell surface expression of pro-apoptotic TRAIL-R on cancer cells and should be considered in clinical trials targeting TRAIL-receptors in cancer patients.
Collapse
Affiliation(s)
- Dhifaf Sarhan
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm S-17176, Sweden.
| | - Padraig D'Arcy
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm S-17176, Sweden.
| | - Andreas Lundqvist
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm S-17176, Sweden.
| |
Collapse
|
306
|
Paul I, Jones JM. Apoptosis block as a barrier to effective therapy in non small cell lung cancer. World J Clin Oncol 2014; 5:588-594. [PMID: 25302163 PMCID: PMC4129524 DOI: 10.5306/wjco.v5.i4.588] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 02/28/2014] [Accepted: 06/11/2014] [Indexed: 02/06/2023] Open
Abstract
Lung cancer, is the most common cause of cancer death in men and second only to breast cancer in women. Currently, the first line therapy of choice is platinum-based combination chemotherapy. A therapeutic plateau has been reached with the prognosis for patients with advanced non-small cell lung cancer (NSCLC) remaining poor. New biomarkers of prognosis as well as new therapies focusing on molecular targets are emerging helping to identify patients who are likely to benefit from therapy. Despite this, drug resistance remains the major cause for treatment failure. In this article we review the role of apoptosis in mediating drug resistance in NSCLC. Better understanding of this fundamental biological process may provide a rationale for overcoming the current therapeutic plateau.
Collapse
|
307
|
Sun T, Zhang YS, Pang B, Hyun DC, Yang M, Xia Y. Engineered nanoparticles for drug delivery in cancer therapy. Angew Chem Int Ed Engl 2014; 53:12320-64. [PMID: 25294565 DOI: 10.1002/anie.201403036] [Citation(s) in RCA: 744] [Impact Index Per Article: 74.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Indexed: 12/18/2022]
Abstract
In medicine, nanotechnology has sparked a rapidly growing interest as it promises to solve a number of issues associated with conventional therapeutic agents, including their poor water solubility (at least, for most anticancer drugs), lack of targeting capability, nonspecific distribution, systemic toxicity, and low therapeutic index. Over the past several decades, remarkable progress has been made in the development and application of engineered nanoparticles to treat cancer more effectively. For example, therapeutic agents have been integrated with nanoparticles engineered with optimal sizes, shapes, and surface properties to increase their solubility, prolong their circulation half-life, improve their biodistribution, and reduce their immunogenicity. Nanoparticles and their payloads have also been favorably delivered into tumors by taking advantage of the pathophysiological conditions, such as the enhanced permeability and retention effect, and the spatial variations in the pH value. Additionally, targeting ligands (e.g., small organic molecules, peptides, antibodies, and nucleic acids) have been added to the surface of nanoparticles to specifically target cancerous cells through selective binding to the receptors overexpressed on their surface. Furthermore, it has been demonstrated that multiple types of therapeutic drugs and/or diagnostic agents (e.g., contrast agents) could be delivered through the same carrier to enable combination therapy with a potential to overcome multidrug resistance, and real-time readout on the treatment efficacy. It is anticipated that precisely engineered nanoparticles will emerge as the next-generation platform for cancer therapy and many other biomedical applications.
Collapse
Affiliation(s)
- Tianmeng Sun
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332 (USA)
| | | | | | | | | | | |
Collapse
|
308
|
Sun T, Zhang YS, Pang B, Hyun DC, Yang M, Xia Y. Maßgeschneiderte Nanopartikel für den Wirkstofftransport in der Krebstherapie. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201403036] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
309
|
Abstract
Natural killer (NK) cells recognize targets stressed by malignant transformation or infection and can be long-lived. They become educated by interacting with major histocompatibility antigen (MHC) class I molecules to gain function to kill targets and produce cytokines. In the clinic, haploidentical NK cells can be adoptively transferred to treat cancer. Persistence and in vivo expansion of NK cells depends on lymphodepleting chemotherapy to make space and induce release of endogenous IL-15. In vivo expansion is also enhanced by cytokine administration but IL-2 has the down side of stimulating CD25hi regulatory T cells (Tregs). Other limitations to NK-cell therapy include poor in vivo survival and lack of specificity. Bispecific or trispecific killer engagers that target CD16 on NK cells to enhance recognition of tumor antigens, and desintegrin and metalloproteinase 17 (ADAM17) inhibition that prevents CD16 shedding after NK-cell activation should promote enhanced killing of cancer with specificity. These are exciting times; more than 35 years after NK cells were initially described, we are exploiting their capacity for clinical therapy.
Collapse
Affiliation(s)
- Veronika Bachanova
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN
| | - Jeffrey S Miller
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN
| |
Collapse
|
310
|
Aplysin sensitizes cancer cells to TRAIL by suppressing P38 MAPK/survivin pathway. Mar Drugs 2014; 12:5072-88. [PMID: 25257790 PMCID: PMC4178493 DOI: 10.3390/md12095072] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/14/2014] [Accepted: 04/15/2014] [Indexed: 01/18/2023] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) is a tumor-selective apoptosis inducer and has been shown to be promising for treating various types of cancers. However, the application of TRAIL is greatly impeded by the resistance of cancer cells to its action. Studies show that overexpression of some critical pro-survival proteins, such as survivin, is responsible for TRAIL resistance. In this study, we found that Aplysin, a brominated compound from marine organisms, was able to restore the sensitivity of cancer cells to TRAIL both in vitro and in vivo. Aplysin was found to enhance the tumor-suppressing capacity of TRAIL on several TRAIL-resistant cancer cell lines. TRAIL-induced apoptosis was also potentiated in A549 and MCF7 cells treated with Aplysin. Survivin downregulation was identified as a mechanism by which Aplysin-mediated TRAIL sensitization of cancer cells. Furthermore, the activation of p38 MAPK was revealed in Aplysin-treated cancer cells, and its inhibitor SB203580 was able to abrogate the promoting effect of Aplysin on the response of cancer cells to TRAIL action, as evidenced by restored survivin expression, elevated cell survival and reduced apoptotic rates. In conclusion, we provided evidence that Aplysin acts as a sensitizer for TRAIL and its effect on p38 MAPK/survivin pathway may partially account for this activity. Considering its low cytotoxicity to normal cells, Aplysin may be a promising agent for cancer treatment in combination with TRAIL.
Collapse
|
311
|
Ullenhag GJ, Al-Attar A, Mukherjee A, Green AR, Ellis IO, Durrant LG. The TRAIL system is over-expressed in breast cancer and FLIP a marker of good prognosis. J Cancer Res Clin Oncol 2014; 141:505-14. [PMID: 25230899 DOI: 10.1007/s00432-014-1822-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 09/01/2014] [Indexed: 02/04/2023]
Abstract
BACKGROUND Breast cancer is the most common cancer in women. The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathway transmits apoptotic signals. Novel anticancer agents that activate this system are in clinical development, including anti-breast cancer. METHODS The tissue microarray technique was applied. We used an array of breast cancer tissues from a large group of patients (>800) to assess the protein expression of TRAIL-R1, TRAIL-R2, the long isoform of FLICE-inhibitory protein and total FLICE-inhibitory protein (FLIP(L) and FLIP(T)). Disease-free survival was examined by Kaplan-Meier estimates and the log-rank test. The independence of prognostic factors was determined by Cox multivariate analysis. RESULTS High intra-tumoral expression of all these proteins of the TRAIL pathway was found. The TRAIL receptors and FLIP(L) were not associated with survival. On univariate analysis, strong FLIP(T) expression was associated with a significantly better survival (p = 0.001). On multivariate analysis using the Cox proportional hazards model, FLIP(T) phenotype was significantly associated with a good prognosis in this series (HR 0.52, 95 % CI 0.35-0.78, p = 0.039). Results indicate that this association is valid for all the biological subtypes of breast cancer. The expression of FLIP(T) was especially high in the luminal subtype, known for its good prognosis. CONCLUSIONS These findings support the use of agonistic TRAIL antibodies and drugs targeting FLIP in breast cancer patients. Over-expression of FLIP(T) but not TRAIL-R1, TRAIL-R2 or FLIP(L) provides stage-independent prognostic information in breast cancer patients. This indicates a clinically less aggressive phenotype.
Collapse
Affiliation(s)
- Gustav J Ullenhag
- Section of Oncology, Department of Radiology, Oncology and Radiation Science, Uppsala University, Uppsala, Sweden,
| | | | | | | | | | | |
Collapse
|
312
|
Bertsch U, Röder C, Kalthoff H, Trauzold A. Compartmentalization of TNF-related apoptosis-inducing ligand (TRAIL) death receptor functions: emerging role of nuclear TRAIL-R2. Cell Death Dis 2014; 5:e1390. [PMID: 25165876 PMCID: PMC4454323 DOI: 10.1038/cddis.2014.351] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/09/2014] [Accepted: 07/10/2014] [Indexed: 01/05/2023]
Abstract
Localized in the plasma membrane, death domain-containing TNF-related apoptosis-inducing ligand (TRAIL) receptors, TRAIL-R1 and TRAIL-R2, induce apoptosis and non-apoptotic signaling when crosslinked by the ligand TRAIL or by agonistic receptor-specific antibodies. Recently, an increasing body of evidence has accumulated that TRAIL receptors are additionally found in noncanonical intracellular locations in a wide range of cell types, preferentially cancer cells. Thus, besides their canonical locations in the plasma membrane and in intracellular membranes of the secretory pathway as well as endosomes and lysosomes, TRAIL receptors may also exist in autophagosomes, in nonmembraneous cytosolic compartment as well as in the nucleus. Such intracellular locations have been mainly regarded as hide-outs for these receptors representing a strategy for cancer cells to resist TRAIL-mediated apoptosis. Recently, a novel function of intracellular TRAIL-R2 has been revealed. When present in the nuclei of tumor cells, TRAIL-R2 inhibits the processing of the primary let-7 miRNA (pri-let-7) via interaction with accessory proteins of the Microprocessor complex. The nuclear TRAIL-R2-driven decrease in mature let-7 enhances the malignancy of cancer cells. This finding represents a new example of nuclear activity of typically plasma membrane-located cytokine and growth factor receptors. Furthermore, this extends the list of nucleic acid targets of the cell surface receptors by pri-miRNA in addition to DNA and mRNA. Here we review the diverse functions of TRAIL-R2 depending on its intracellular localization and we particularly discuss the nuclear TRAIL-R2 (nTRAIL-R2) function in the context of known nuclear activities of other normally plasma membrane-localized receptors.
Collapse
Affiliation(s)
- U Bertsch
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel D-24105, Germany
| | - C Röder
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel D-24105, Germany
| | - H Kalthoff
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel D-24105, Germany
| | - A Trauzold
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel D-24105, Germany
| |
Collapse
|
313
|
Esophageal cancer-selective expression of TRAIL mediated by MREs of miR-143 and miR-122. Tumour Biol 2014; 35:5787-95. [PMID: 24659424 DOI: 10.1007/s13277-014-1768-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 02/17/2014] [Indexed: 10/25/2022] Open
Abstract
Esophageal cancer is one of the most common digestive system neoplasms and has a quite poor prognosis. TNF-related apoptosis-inducing ligand (TRAIL) induces the apoptosis in a wide range of cancer cells including esophageal cancers. However, TRAIL also activates apoptotic pathway in normal cells. To improve the specificity of TRAIL action, we employed the microRNA (miRNA) response elements (MREs) of miR-143 and miR-122 to restrict TRAIL expression mediated by an adenoviral vector (Ad-TRAIL-143-122) in esophageal cancer cells. The experiments showed that Ad-TRAIL-143-122 was able to highly express TRAIL in esophageal cancer cells, but not normal cells. The selective TRAIL expression also led to selective apoptosis in esophageal cancer cells. Ad-TRAIL-143-122 greatly reduced the viability of esophageal cancer cells without cytotoxicity to normal cells. In mice, Ad-TRAIL-143-122 suppressed the growth of esophageal cancer xenografts and protected liver from TRAIL-induced toxicity. In this study, we constructed a biologic vector that can express exogenous genes in a tumor-specific manner. This strategy can simultaneously treat cancer and prevent hepatoxicity and thus may be a promising way for esophageal cancer treatment.
Collapse
|
314
|
Xiao X, Liao X, Qiu S, Liu Z, Du B, Xu S. Synthesis, cytotoxicity and apoptosis induction in human tumor cells by galaxamide and its analogues [corrected]. Mar Drugs 2014; 12:4521-38. [PMID: 25231922 PMCID: PMC4145329 DOI: 10.3390/md12084521] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 07/05/2014] [Accepted: 07/29/2014] [Indexed: 11/16/2022] Open
Abstract
Our previous study reported that galaxamide, which is a cyclo-pentapeptide containing five leucines that was extracted from Galaxaura filamentosa, displayed remarkable anticancer cytotoxicity. This novel cyclo-peptide provided a new skeleton for the structural modifications used in finding new drugs with better anticancer properties. In this study, five analogues were synthesized based on changing the number of d/l amino acids by adding a new amino acid, phenylalanine. Galaxamide and five of its analogues were evaluated through MTT assays to examine their cytotoxic activities. We found that modified analogue 5, which is referred to as A5, displayed broad spectrum cytotoxic activity toward every cell line tested; in addition, the IC50 of A5 was lower than that of galaxamide and the other analogues. Furthermore, we used flow cytometry and western blot assays to investigate whether galaxamide and A5 could induce cancer cell apoptosis. The flow cytometric studies showed that HepG2 cells treated with different concentrations of galaxamide or A5 over 72 h displayed significant and dose-dependent increases in the percentages of early-stage apoptotic cells. Western blotting revealed that both compounds induce caspase-dependent apoptosis in HepG2 cells through a mitochondria-mediated pathway. The results demonstrate that galaxamide and its analogues have potential applications as clinical anticancer drugs.
Collapse
Affiliation(s)
- Xi Xiao
- Department of Chemistry, Life Science School, Jinan University, Guangzhou 510632, China.
| | - Xiaojian Liao
- Department of Chemistry, Life Science School, Jinan University, Guangzhou 510632, China.
| | - Shaoling Qiu
- Department of Chemistry, Life Science School, Jinan University, Guangzhou 510632, China.
| | - Zihao Liu
- Department of Pathology, Medical School, Jinan University, Guangzhou 510632, China.
| | - Bin Du
- Department of Pathology, Medical School, Jinan University, Guangzhou 510632, China.
| | - Shihai Xu
- Department of Chemistry, Life Science School, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
315
|
Graves JD, Kordich JJ, Huang TH, Piasecki J, Bush TL, Sullivan T, Foltz IN, Chang W, Douangpanya H, Dang T, O'Neill JW, Mallari R, Zhao X, Branstetter DG, Rossi JM, Long AM, Huang X, Holland PM. Apo2L/TRAIL and the death receptor 5 agonist antibody AMG 655 cooperate to promote receptor clustering and antitumor activity. Cancer Cell 2014; 26:177-89. [PMID: 25043603 DOI: 10.1016/j.ccr.2014.04.028] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 02/28/2014] [Accepted: 04/30/2014] [Indexed: 12/15/2022]
Abstract
Death receptor agonist therapies have exhibited limited clinical benefit to date. Investigations into why Apo2L/TRAIL and AMG 655 preclinical data were not predictive of clinical response revealed that coadministration of Apo2L/TRAIL with AMG 655 leads to increased antitumor activity in vitro and in vivo. The combination of Apo2L/TRAIL and AMG 655 results in enhanced signaling and can sensitize Apo2L/TRAIL-resistant cells. Structure determination of the Apo2L/TRAIL-DR5-AMG 655 ternary complex illustrates how higher order clustering of DR5 is achieved when both agents are combined. Enhanced agonism generated by combining Apo2L/TRAIL and AMG 655 provides insight into the limited efficacy observed in previous clinical trials and suggests testable hypotheses to reconsider death receptor agonism as a therapeutic strategy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Cell Survival
- Crystallography, X-Ray
- Drug Resistance, Neoplasm
- Drug Synergism
- Humans
- Mice
- Models, Molecular
- Protein Multimerization
- Protein Structure, Quaternary
- Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors
- Receptors, TNF-Related Apoptosis-Inducing Ligand/chemistry
- Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
- Signal Transduction
- TNF-Related Apoptosis-Inducing Ligand/chemistry
- TNF-Related Apoptosis-Inducing Ligand/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
| | | | - Tzu-Hsuan Huang
- Therapeutic Innovation Unit, Amgen Inc., Cambridge, MA 02142, USA
| | - Julia Piasecki
- Therapeutic Innovation Unit, Amgen Inc., Seattle, WA 98119, USA
| | - Tammy L Bush
- Therapeutic Innovation Unit, Amgen Inc., Cambridge, MA 02142, USA
| | - Timothy Sullivan
- Therapeutic Innovation Unit, Amgen Inc., Cambridge, MA 02142, USA
| | - Ian N Foltz
- Department of Biologic Discovery, Amgen British Columbia, Burnaby, BC V5A 1V7, Canada
| | - Wesley Chang
- Department of Clinical Immunology, Amgen Inc., South San Francisco, CA 94080, USA
| | | | - Thu Dang
- Therapeutic Innovation Unit, Amgen Inc., Cambridge, MA 02142, USA
| | - Jason W O'Neill
- Department of Biologic Optimization, Amgen Inc., Seattle, WA 98119, USA
| | - Rommel Mallari
- Department of Molecular Structure and Characterization, Amgen, South San Francisco, CA, 94080, USA
| | - Xiaoning Zhao
- Department of Molecular Structure and Characterization, Amgen, South San Francisco, CA, 94080, USA
| | | | - John M Rossi
- Department of Molecular Sciences and Computational Biology, Amgen Inc., Thousand Oaks, CA 91320, USA
| | - Alexander M Long
- Department of Molecular Structure and Characterization, Amgen Inc., Cambridge, MA 02142, USA
| | - Xin Huang
- Department of Molecular Structure and Characterization, Amgen Inc., Cambridge, MA 02142, USA
| | - Pamela M Holland
- Therapeutic Innovation Unit, Amgen Inc., Cambridge, MA 02142, USA.
| |
Collapse
|
316
|
Wu G, Ji Z, Li H, Lei Y, Jin X, Yu Y, Sun M. Selective TRAIL-induced cytotoxicity to lung cancer cells mediated by miRNA response elements. Cell Biochem Funct 2014; 32:547-56. [PMID: 25132116 DOI: 10.1002/cbf.3042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 05/14/2014] [Accepted: 05/28/2014] [Indexed: 12/18/2022]
Abstract
Lung cancer is among the most common cancers, and the current therapeutic strategies are still inefficient in most cases. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising biological agent for cancer treatment because of its potent pro-apoptotic effect on cancer cells. However, TRAIL also induces apoptosis in normal cells and therefore may cause toxicity to normal tissues if clinically applied. To address this issue, we inserted microRNA response elements (MREs) of miR-133a, miR-137 and miR-449a, which are all underexpressed in lung cancer cells, into an adenoviral vector to regulate TRAIL expression. This MRE-regulated vector (Ad-TRAIL-MRE) was able to express TRAIL in a lung-cancer-specific fashion. No TRAIL expression was detected in normal cells. Consistently, Ad-TRAIL-MRE exerted cytotoxicity to lung cancer cells, rather than normal cells, perhaps via inducing selective apoptosis. The selective TRAIL-mediated growth-inhibiting effect was further confirmed in a tumour xenograft model. Also, Ad-TRAIL-MRE only resulted in very low hepatotoxicity when applied. Collectively, we generated a novel TRAIL-expressing adenoviral vector that was regulated by MREs. This strategy permits TRAIL expression in a lung-cancer-specific manner and is worth further studying for clinical trials.
Collapse
Affiliation(s)
- Guodong Wu
- Department of Cardiovascular, Regenerative Medicine and Tissue Engineering, First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | | | | | | | | | | | | |
Collapse
|
317
|
Shin EA, Sohn EJ, Won G, Choi JU, Jeong M, Kim B, Kim MJ, Kim SH. Upregulation of microRNA135a-3p and death receptor 5 plays a critical role in Tanshinone I sensitized prostate cancer cells to TRAIL induced apoptosis. Oncotarget 2014; 5:5624-36. [PMID: 25015549 PMCID: PMC4170628 DOI: 10.18632/oncotarget.2152] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 06/29/2014] [Indexed: 01/24/2023] Open
Abstract
Though tumor necrosis factor related apoptosis inducing ligand (TRAIL) has been used as a potent anticancer agent, TRAIL resistance is a hot-issue in cancer therapy. We investigated the antitumor mechanism of Tanshinone I to sensitize prostate cancer cells to TRAIL. Comibination of Tanshinone I and TRAIL exerted synergistic cytotoxicity, increased cleaved PARP, sub G1 population, the number of TUNELpositive cells, activated caspase 8, 9 and ROS production in PC-3 and DU145 cells. Of note, combination of Tanshinone I and TRAIL enhanced the protein expression of death receptor 5 (DR5) and attenuated anti-apoptotic proteins. RT-PCR and RT-qPCR analyses confirmed that co-treatment of Tanshinone I and TRAIL up-regulated DR5 and microRNA 135a-3p at mRNA level or activity of DR5 promoter and attenuated phosphorylation of extracellular signal regulated kinases in PC-3. Conversely, the silencing of DR5 blocked the increased cytotoxicity, sub G1 population and PARP cleavages induced by co-treatment of Tanshinone I and TRAIL. Interestingly, miR135a-3p mimic enhanced DR5 at mRNA, increased PARP cleavage, Bax and the number of TUNEL positive cells in Tanshinone I and TRAIL cotreated PC-3. Overall, our findings suggest that Tanshinone I enhances TRAIL mediated apoptosis via upregulation of miR135a-3p mediated DR5 in prostate cancer cells as a potent TRAIL sensitizer.
Collapse
Affiliation(s)
- Eun Ah Shin
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea
| | - Eun Jung Sohn
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea
| | - Gunho Won
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea
| | - Jeong-Un Choi
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea
| | - Myongsuk Jeong
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea
| | - Bonglee Kim
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea
| | - Min-Jeong Kim
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea
| | - Sung-Hoon Kim
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
318
|
Brunetti G, Oranger A, Carbone C, Mori G, Sardone FR, Mori C, Celi M, Faienza MF, Tarantino U, Zallone A, Grano M, Colucci S. Osteoblasts display different responsiveness to TRAIL-induced apoptosis during their differentiation process. Cell Biochem Biophys 2014; 67:1127-36. [PMID: 23677859 DOI: 10.1007/s12013-013-9616-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Apoptosis can occur throughout the life span of osteoblasts (OBs), beginning from the early stages of differentiation and continuing throughout all stages of their working life. Here, we investigated the effects of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) on normal human OBs showing for the first time that the expression of TRAIL receptors is modulated during OB differentiation. In particular, the TRAIL receptor ratio was in favor of the deaths because of the low expression of DcR2 in undifferentiated OBs, differently it was shifted toward the decoys in differentiated ones. Undifferentiated OBs treated with TRAIL showed reduced cell viability, whereas differentiated OBs displayed TRAIL resistance. The OB sensitiveness to TRAIL was due to the up-regulation of DR5 and the down-regulation of DcR2. The main death receptor involved in TRAIL-reduced OB viability was DR5 as demonstrated by the rescue of cell viability observed in the presence of anti-DR5 neutralizing antibody. Besides the ratio of TRAIL receptors, the sensitivity of undifferentiated OBs to TRAIL-cytotoxic effect was also associated with low mRNA levels of intracellular anti-apoptotic proteins, such as cFLIP, the activation of caspase-8 and -3, as well as the DNA fragmentation. This study suggests that apoptotic effect exerted by TRAIL/TRAIL-receptor system on normal human OB is strictly dependent upon cell differentiation status.
Collapse
Affiliation(s)
- Giacomina Brunetti
- Section of Human Anatomy and Histology - R. Amprino, Department of Basic Medical Sciences, Neuroscience and Sense Organs, Medical School, University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
319
|
Schlegel CR, Fonseca AV, Stöcker S, Georgiou ML, Misterek MB, Munro CE, Carmo CR, Seckl MJ, Costa-Pereira AP. DAPK2 is a novel modulator of TRAIL-induced apoptosis. Cell Death Differ 2014; 21:1780-91. [PMID: 25012503 DOI: 10.1038/cdd.2014.93] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 05/27/2014] [Accepted: 05/28/2014] [Indexed: 12/22/2022] Open
Abstract
Targeting molecules involved in TRAIL-mediated signalling has been hailed by many as a potential magic bullet to kill cancer cells efficiently, with little side effects on normal cells. Indeed, initial clinical trials showed that antibodies against TRAIL receptors, death receptor (DR)4 and DR5, are well tolerated by cancer patients. Despite efficacy issues in the clinical setting, novel approaches to trigger TRAIL-mediated apoptosis are being developed and its clinical potential is being reappraised. Unfortunately, as observed with other cancer therapies, many patients develop resistance to TRAIL-induced apoptosis and there is thus impetuous for identifying additional resistance mechanisms that may be targetable and usable in combination therapies. Here, we show that the death-associated protein kinase 2 (DAPK2) is a modulator of TRAIL signalling. Genetic ablation of DAPK2 using RNA interference causes phosphorylation of NF-κB and its transcriptional activity in several cancer cell lines. This then leads to the induction of a variety of NF-κB target genes, which include proapoptotic DR4 and DR5. DR4 and DR5 protein expression is correspondingly increased on the cell surface and this leads to the sensitisation of resistant cells to TRAIL-induced killing, in a p53-independent manner. As DAPK2 is a kinase, it is imminently druggable, and our data thus offer a novel avenue to overcome TRAIL resistance in the clinic.
Collapse
Affiliation(s)
- C R Schlegel
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, Hammersmith Hospital, Hammersmith Hospital Campus, ICTEM, Du Cane Road, London W12 0NN, UK
| | - A-V Fonseca
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, Hammersmith Hospital, Hammersmith Hospital Campus, ICTEM, Du Cane Road, London W12 0NN, UK
| | - S Stöcker
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, Hammersmith Hospital, Hammersmith Hospital Campus, ICTEM, Du Cane Road, London W12 0NN, UK
| | - M L Georgiou
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, Hammersmith Hospital, Hammersmith Hospital Campus, ICTEM, Du Cane Road, London W12 0NN, UK
| | - M B Misterek
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, Hammersmith Hospital, Hammersmith Hospital Campus, ICTEM, Du Cane Road, London W12 0NN, UK
| | - C E Munro
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, Hammersmith Hospital, Hammersmith Hospital Campus, ICTEM, Du Cane Road, London W12 0NN, UK
| | - C R Carmo
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, Hammersmith Hospital, Hammersmith Hospital Campus, ICTEM, Du Cane Road, London W12 0NN, UK
| | - M J Seckl
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, Hammersmith Hospital, Hammersmith Hospital Campus, ICTEM, Du Cane Road, London W12 0NN, UK
| | - A P Costa-Pereira
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, Hammersmith Hospital, Hammersmith Hospital Campus, ICTEM, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
320
|
Liu YJ, Lin YC, Lee JC, Kuo SC, Ho CT, Huang LJ, Kuo DH, Way TD. CCT327 enhances TRAIL-induced apoptosis through the induction of death receptors and downregulation of cell survival proteins in TRAIL-resistant human leukemia cells. Oncol Rep 2014; 32:1257-64. [PMID: 25017974 DOI: 10.3892/or.2014.3317] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 05/20/2014] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor-related apoptosis‑inducing ligand (TRAIL) has potential application in cancer therapy and it has the ability to selectively kill cancer cells without affecting normal cells. However, the development of resistance to TRAIL in cancer cells cannot be avoided. This study investigated the effects of 2-(5-methylselenophen‑2‑yl)‑6,7‑methylenedioxyquinolin‑4-one (CCT327), an analogue of quinolin-4-one, on the sensitization of cancer cells to TRAIL and on TRAIL‑induced apoptosis in TRAIL‑resistance human leukemia cells (HL60‑TR). We found that CCT327 enhanced TRAIL‑induced apoptosis through upregulation of death receptors DR4 and DR5. In addition to upregulating DRs (death receptors), CCT327 suppressed the expression of decoy receptor DcR1 and DcR2. CCT327 significantly downregulated the expression of FLICE inhibitory protein (cFLIP) and other antiapoptotic proteins. We also demonstrated that CCT327 could activate p38 and JNK. Moreover, CCT327-induced induction of DR5 and DR4 was mediated by reactive oxygen species (ROS), and N-acetylcysteine (NAC) blocked the induction of DRs by CCT327. Taken together, these results showed that CCT327 combined with TRAIL treatment may provide an effective therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Yan-Jin Liu
- Graduate Institute of Pharmaceutical Chemistry, College of Pharmacy, China Medical University, Taichung, Taiwan, R.O.C
| | - Ying-Chao Lin
- Division of Neurosurgery, Buddhist Tzu Chi General Hospital, Taichung Branch, Taichung, Taiwan, R.O.C
| | - Jang-Chang Lee
- Department of Pharmacy, China Medical University, Taichung, Taiwan, R.O.C
| | - Sheng-Chu Kuo
- Graduate Institute of Pharmaceutical Chemistry, College of Pharmacy, China Medical University, Taichung, Taiwan, R.O.C
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Li-Jiau Huang
- Graduate Institute of Pharmaceutical Chemistry, College of Pharmacy, China Medical University, Taichung, Taiwan, R.O.C
| | - Daih-Huang Kuo
- Graduate Institute of Pharmaceutical Technology, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan, R.O.C
| | - Tzong-Der Way
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan, R.O.C
| |
Collapse
|
321
|
Synergistic Antitumor Activity of Recombinant Human Apo2L/Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) in Combination with Carboplatin and Pemetrexed in Malignant Pleural Mesothelioma. J Thorac Oncol 2014; 9:1008-1017. [DOI: 10.1097/jto.0000000000000198] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
322
|
Warat M, Szliszka E, Korzonek-Szlacheta I, Król W, Czuba ZP. Chrysin, apigenin and acacetin inhibit tumor necrosis factor-related apoptosis-inducing ligand receptor-1 (TRAIL-R1) on activated RAW264.7 macrophages. Int J Mol Sci 2014; 15:11510-22. [PMID: 24979133 PMCID: PMC4139796 DOI: 10.3390/ijms150711510] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 05/15/2014] [Accepted: 06/10/2014] [Indexed: 12/13/2022] Open
Abstract
Expression level of Tumor Necrosis Factor—related apoptosis—inducing ligand (TRAIL) receptors is one of the most important factors of TRAIL-mediated apoptosis in cancer cells. We here report for the first time data concerning TRAIL-R1 and TRAIL-R2 receptor expression on RAW264.7 macrophages. Three substances belonging to flavones: chrysin, apigenin and acacetin which differ from their substituents at the 4' position in the phenyl ring were used in assays because of the variety of biological activities (e.g., anticancer activity) of the polyphenol compounds. The expression of TRAIL-R1 and TRAIL-R2 death receptors on non-stimulated and LPS (lipopolysaccharide)-stimulated macrophages was determined using flow cytometry. We demonstrate that RAW264.7 macrophages exhibit TRAIL-R1 surface expression and that the tested compounds: chrysin, apigenin and acacetin can inhibit TRAIL-R1 death receptor expression level on macrophages.
Collapse
Affiliation(s)
- Monika Warat
- Chair and Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Jordana 19, 41-808 Zabrze, Poland.
| | - Ewelina Szliszka
- Chair and Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Jordana 19, 41-808 Zabrze, Poland.
| | - Ilona Korzonek-Szlacheta
- Department of Toxicology and Health Protection, Toxicology and Drug Addiction Division, Medical University of Silesia in Katowice, Medyków 18, 40-752 Katowice, Poland.
| | - Wojciech Król
- Chair and Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Jordana 19, 41-808 Zabrze, Poland.
| | - Zenon P Czuba
- Chair and Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Jordana 19, 41-808 Zabrze, Poland.
| |
Collapse
|
323
|
Lemke J, von Karstedt S, Zinngrebe J, Walczak H. Getting TRAIL back on track for cancer therapy. Cell Death Differ 2014; 21:1350-64. [PMID: 24948009 PMCID: PMC4131183 DOI: 10.1038/cdd.2014.81] [Citation(s) in RCA: 369] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/14/2014] [Accepted: 04/15/2014] [Indexed: 02/07/2023] Open
Abstract
Unlike other members of the TNF superfamily, the TNF-related apoptosis-inducing ligand (TRAIL, also known as Apo2L) possesses the unique capacity to induce apoptosis selectively in cancer cells in vitro and in vivo. This exciting discovery provided the basis for the development of TRAIL-receptor agonists (TRAs), which have demonstrated robust anticancer activity in a number of preclinical studies. Subsequently initiated clinical trials testing TRAs demonstrated, on the one hand, broad tolerability but revealed, on the other, that therapeutic benefit was rather limited. Several factors that are likely to account for TRAs' sobering clinical performance have since been identified. First, because of initial concerns over potential hepatotoxicity, TRAs with relatively weak agonistic activity were selected to enter clinical trials. Second, although TRAIL can induce apoptosis in several cancer cell lines, it has now emerged that many others, and importantly, most primary cancer cells are resistant to TRAIL monotherapy. Third, so far patients enrolled in TRA-employing clinical trials were not selected for likelihood of benefitting from a TRA-comprising therapy on the basis of a valid(ated) biomarker. This review summarizes and discusses the results achieved so far in TRA-employing clinical trials in the light of these three shortcomings. By integrating recent insight on apoptotic and non-apoptotic TRAIL signaling in cancer cells, we propose approaches to introduce novel, revised TRAIL-based therapeutic concepts into the cancer clinic. These include (i) the use of recently developed highly active TRAs, (ii) the addition of efficient, but cancer-cell-selective TRAIL-sensitizing agents to overcome TRAIL resistance and (iii) employing proteomic profiling to uncover resistance mechanisms. We envisage that this shall enable the design of effective TRA-comprising therapeutic concepts for individual cancer patients in the future.
Collapse
Affiliation(s)
- J Lemke
- 1] Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK [2] Clinic of General and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - S von Karstedt
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - J Zinngrebe
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - H Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| |
Collapse
|
324
|
Starkey MR, Nguyen DH, Essilfie AT, Kim RY, Hatchwell LM, Collison AM, Yagita H, Foster PS, Horvat JC, Mattes J, Hansbro PM. Tumor necrosis factor-related apoptosis-inducing ligand translates neonatal respiratory infection into chronic lung disease. Mucosal Immunol 2014; 7:478-88. [PMID: 24045576 DOI: 10.1038/mi.2013.65] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 07/24/2013] [Accepted: 08/13/2013] [Indexed: 02/04/2023]
Abstract
Respiratory infections in early life can lead to chronic respiratory disease. Chlamydia infections are common causes of respiratory disease, particularly pneumonia in neonates, and are linked to permanent reductions in pulmonary function and the induction of asthma. However, the immune responses that protect against early-life infection and the mechanisms that lead to chronic lung disease are incompletely understood. Here we identify novel roles for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in promoting Chlamydia respiratory infection-induced pathology in early life, and subsequent chronic lung disease. By infecting TRAIL-deficient neonatal mice and using neutralizing antibodies against this factor and its receptors in wild-type mice, we demonstrate that TRAIL is critical in promoting infection-induced histopathology, inflammation, and mucus hypersecretion, as well as subsequent alveolar enlargement and impaired lung function. This suggests that therapeutic agents that target TRAIL or its receptors may be effective treatments for early-life respiratory infections and associated chronic lung disease.
Collapse
Affiliation(s)
- M R Starkey
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - D H Nguyen
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - A T Essilfie
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - R Y Kim
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - L M Hatchwell
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - A M Collison
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - H Yagita
- Department of Immunology, Juntendo University School of Medicine, Hongo, Bunkyo-ku, Tokyo, Japan
| | - P S Foster
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - J C Horvat
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - J Mattes
- 1] Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia [2] Pediatric Respiratory and Sleep Medicine Unit, Newcastle Children's Hospital, Kaleidoscope, New Lambton Heights, Newcastle, New South Wales, Australia
| | - P M Hansbro
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| |
Collapse
|
325
|
Fouqué A, Debure L, Legembre P. The CD95/CD95L signaling pathway: a role in carcinogenesis. Biochim Biophys Acta Rev Cancer 2014; 1846:130-41. [PMID: 24780723 DOI: 10.1016/j.bbcan.2014.04.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/17/2014] [Accepted: 04/19/2014] [Indexed: 11/18/2022]
Abstract
Apoptosis is a fundamental process that contributes to tissue homeostasis, immune responses, and development. The receptor CD95, also called Fas, is a member of the tumor necrosis factor receptor (TNF-R) superfamily. Its cognate ligand, CD95L, is implicated in immune homeostasis and immune surveillance, and various lineages of malignant cells exhibit loss-of-function mutations in this pathway; therefore, CD95 was initially classified as a tumor suppressor gene. However, more recent data indicate that in different pathophysiological contexts, this receptor can transmit non-apoptotic signals, promote inflammation, and contribute to carcinogenesis. A comparison with the initial molecular events of the TNF-R signaling pathway leading to non-apoptotic, apoptotic, and necrotic pathways reveals that CD95 is probably using different molecular mechanisms to transmit its non-apoptotic signals (NF-κB, MAPK, and PI3K). As discussed in this review, the molecular process by which the receptor switches from an apoptotic function to an inflammatory role is unknown. More importantly, the biological functions of these signals remain elusive.
Collapse
Affiliation(s)
- Amélie Fouqué
- Université Rennes-1, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; INSERM U1085, IRSET, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; Equipe Labellisée Ligue Contre Le Cancer "Death Receptors and Tumor Escape", 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; Centre Eugène Marquis, rue bataille Flandres Dunkerque, Rennes, France
| | - Laure Debure
- Université Rennes-1, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; INSERM U1085, IRSET, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; Equipe Labellisée Ligue Contre Le Cancer "Death Receptors and Tumor Escape", 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; Centre Eugène Marquis, rue bataille Flandres Dunkerque, Rennes, France
| | - Patrick Legembre
- Université Rennes-1, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; INSERM U1085, IRSET, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; Equipe Labellisée Ligue Contre Le Cancer "Death Receptors and Tumor Escape", 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; Centre Eugène Marquis, rue bataille Flandres Dunkerque, Rennes, France.
| |
Collapse
|
326
|
Du W, Uslar L, Sevala S, Shah K. Targeting c-Met receptor overcomes TRAIL-resistance in brain tumors. PLoS One 2014; 9:e95490. [PMID: 24748276 PMCID: PMC3991662 DOI: 10.1371/journal.pone.0095490] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 03/27/2014] [Indexed: 12/22/2022] Open
Abstract
Tumor necrosis factor related apoptosis-inducing ligand (TRAIL) induced apoptosis specifically in tumor cells. However, with approximately half of all known tumor lines being resistant to TRAIL, the identification of TRAIL sensitizers and their mechanism of action become critical to broadly use TRAIL as a therapeutic agent. In this study, we explored whether c-Met protein contributes to TRAIL sensitivity. We found a direct correlation between the c-Met expression level and TRAIL resistance. We show that the knock down c-Met protein, but not inhibition, sensitized brain tumor cells to TRAIL-mediated apoptosis by interrupting the interaction between c-Met and TRAIL cognate death receptor (DR) 5. This interruption greatly induces the formation of death-inducing signaling complex (DISC) and subsequent downstream apoptosis signaling. Using intracranially implanted brain tumor cells and stem cell (SC) lines engineered with different combinations of fluorescent and bioluminescent proteins, we show that SC expressing a potent and secretable TRAIL (S-TRAIL) have a significant anti-tumor effect in mice bearing c-Met knock down of TRAIL-resistant brain tumors. To our best knowledge, this is the first study that demonstrates c-Met contributes to TRAIL sensitivity of brain tumor cells and has implications for developing effective therapies for brain tumor patients.
Collapse
Affiliation(s)
- Wanlu Du
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Liubov Uslar
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sindhura Sevala
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Khalid Shah
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
327
|
Lin YC, Richburg JH. Characterization of the role of tumor necrosis factor apoptosis inducing ligand (TRAIL) in spermatogenesis through the evaluation of trail gene-deficient mice. PLoS One 2014; 9:e93926. [PMID: 24736722 PMCID: PMC3988040 DOI: 10.1371/journal.pone.0093926] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 03/10/2014] [Indexed: 11/19/2022] Open
Abstract
TRAIL (TNFSF10/Apo2L) is a member of the tumor necrosis factor (TNF) superfamily of proteins and is expressed in human and rodent testis. Although the functional role of TRAIL in spermatogenesis is not known, TRAIL is recognized to induce apoptosis via binding to its cognate receptors; DR4 (TRAIL-R1/TNFRSF10A) and DR5 (TRAIL-R2/TNFRSF10B). Here, we utilize Trail gene-deficient (Trail-/-) mice to evaluate the role of TRAIL in spermatogenesis by measuring testis weight, germ cell apoptosis, and spermatid head count at postnatal day (PND) 28 (pubertal) and PND 56 (adult). Trail-/- mice have significantly reduced testis to body weight ratios as compared to wild-type C57BL/6J at both ages. Also, Trail-/- mice (PND 28) show a dramatic increase in basal germ cell apoptotic index (AI, 16.77) as compared to C57BL/6J (3.5). In the testis of adult C57BL/6J mice, the AI was lower than in PND 28 C57BL/6J mice (2.2). However, in adult Trail-/- mice, the AI was still higher than that of controls (9.0); indicating a relative high incidence of germ cell apoptosis. Expression of cleaved caspase-8 (CC8) and cleaved caspase-9 (CC9) (markers of the extrinsic and intrinsic apoptotic pathway, respectively) revealed a two-fold increase in the activity of both pathways in adult Trail-/- mice compared to C57BL/6J. Spermatid head counts in adult Trail-/- mice were dramatically reduced by 54% compared to C57BL/6J, indicating these animals suffer a marked decline in the production of mature spermatozoa. Taken together, these findings indicate that TRAIL is an important signaling molecule for maintaining germ cell homeostasis and functional spermatogenesis in the testis.
Collapse
Affiliation(s)
- Yi-Chen Lin
- Division of Pharmacology and Toxicology; Center for Molecular and Cellular Toxicology, College of Pharmacy; The University of Texas at Austin, Austin, Texas, United States of America
| | - John H. Richburg
- Division of Pharmacology and Toxicology; Center for Molecular and Cellular Toxicology, College of Pharmacy; The University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
328
|
Yi L, Zongyuan Y, Cheng G, Lingyun Z, Guilian Y, Wei G. Quercetin enhances apoptotic effect of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in ovarian cancer cells through reactive oxygen species (ROS) mediated CCAAT enhancer-binding protein homologous protein (CHOP)-death receptor 5 pathway. Cancer Sci 2014; 105:520-7. [PMID: 24612139 PMCID: PMC4317845 DOI: 10.1111/cas.12395] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/10/2014] [Accepted: 03/05/2014] [Indexed: 12/11/2022] Open
Abstract
Although tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has shown efficacy in a phase 2 clinical trial, development of resistance to TRAIL by tumor cells is a major roadblock. We investigated whether quercetin, a flavonoid, can sensitize human ovarian cancer cells to TRAIL. Results indicate that quercetin sensitized cancer cells to TRAIL. The quercetin induced expression of death receptor DR5 but did not affect expression of DR4 in cancer cells. The induction of DR5 was mediated through activation of JNK and through upregulation of a transcription factor CCAAT enhancer-binding protein homologous protein (CHOP); as silencing of these signaling molecules abrogated the effect of quercetin. Upregulation of DR5 was mediated through the generation of reactive oxygen species (ROS), as ROS scavengers reduced the effect of quercetin on JNK activation, CHOP upregulation, DR induction, TRAIL sensitization, downregulated the expression of cell survival proteins and upregulated the proapoptotic proteins. Furthermore, quercetin enhances TRAIL mediated inhibition of tumor growth of human SKOV-3 xenograft was associated with induction of apoptosis, activation of caspase-3, CHOP and DR5. Overall, our data suggest that quercetin enhances apoptotic death of ovarian cancer cells to TRAIL through upregulation of CHOP-induced DR5 expression following ROS mediated endoplasmic reticulum-stress.
Collapse
Affiliation(s)
- Liu Yi
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | |
Collapse
|
329
|
Deutsch AJA, Rinner B, Wenzl K, Pichler M, Troppan K, Steinbauer E, Schwarzenbacher D, Reitter S, Feichtinger J, Tierling S, Prokesch A, Scheideler M, Krogsdam A, Thallinger GG, Schaider H, Beham-Schmid C, Neumeister P. NR4A1-mediated apoptosis suppresses lymphomagenesis and is associated with a favorable cancer-specific survival in patients with aggressive B-cell lymphomas. Blood 2014; 123:2367-77. [PMID: 24553175 DOI: 10.1182/blood-2013-08-518878] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
NR4A1 (Nur77) and NR4A3 (Nor-1) function as tumor suppressor genes as demonstrated by the rapid development of acute myeloid leukemia in the NR4A1 and NR4A3 knockout mouse. The aim of our study was to investigate NR4A1 and NR4A3 expression and function in lymphoid malignancies. We found a vastly reduced expression of NR4A1 and NR4A3 in chronic lymphocytic B-cell leukemia (71%), in follicular lymphoma (FL, 70%), and in diffuse large B-cell lymphoma (DLBCL, 74%). In aggressive lymphomas (DLBCL and FL grade 3), low NR4A1 expression was significantly associated with a non-germinal center B-cell subtype and with poor overall survival. To investigate the function of NR4A1 in lymphomas, we overexpressed NR4A1 in several lymphoma cell lines. Overexpression of NR4A1 led to a higher proportion of lymphoma cells undergoing apoptosis. To test the tumor suppressor function of NR4A1 in vivo, the stable lentiviral-transduced SuDHL4 lymphoma cell line harboring an inducible NR4A1 construct was further investigated in xenografts. Induction of NR4A1 abrogated tumor growth in the NSG mice, in contrast to vector controls, which formed massive tumors. Our data suggest that NR4A1 has proapoptotic functions in aggressive lymphoma cells and define NR4A1 as a novel gene with tumor suppressor properties involved in lymphomagenesis.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Blotting, Western
- Cell Line, Tumor
- DNA-Binding Proteins/genetics
- Heterografts
- Humans
- Immunohistochemistry
- Kaplan-Meier Estimate
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/mortality
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Proportional Hazards Models
- Receptors, Steroid/genetics
- Receptors, Thyroid Hormone/genetics
Collapse
|
330
|
Chen W, Tang WR, Zhang M, Chang K, Wei YL. Association of DR4 (TRAIL-R1) Polymorphisms with Cancer Risk in Caucasians: an Updated Meta-analysis. Asian Pac J Cancer Prev 2014; 15:2889-92. [DOI: 10.7314/apjcp.2014.15.6.2889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
331
|
Rai R, Sharma KL, Sharma S, Misra S, Kumar A, Mittal B. Death receptor (DR4) haplotypes are associated with increased susceptibility of gallbladder carcinoma in north Indian population. PLoS One 2014; 9:e90264. [PMID: 24587306 PMCID: PMC3938657 DOI: 10.1371/journal.pone.0090264] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/27/2014] [Indexed: 01/01/2023] Open
Abstract
Background and Aim Defective apoptosis is a hallmark of cancer development and progression. Death receptors (DR4, FAS) and their ligands (TRAIL, FASL) are thought to mediate the major extrinsic apoptotic pathway in the cell. SNPs in these genes may lead to defective apoptosis. Hence, the present study aimed to investigate the association of functional SNPs of DR4 (rs20575, rs20576 and rs6557634), FAS (rs2234767) and FASL (rs763110) with gallbladder cancer (GBC) risk. Methods This case-control study included 400 GBC and 246 healthy controls (HC). Genotyping was carried out by Taqman genotyping assays. Statistical analysis was performed by using SPSS ver16. Meta-analysis was performed using Comprehensive Meta-analysis software (Version 2.0, BIOSTAT, Englewood, NJ) to systematically summarize the possible association of SNP with cancer risk. Functional prediction of these variants was carried out using Bioinformatics tools (FAST-SNP, F-SNP). False discovery rate (FDR test) was used in multiple comparisons. Results The DR4 Crs20575Ars20576Ars6557634, Grs20575Ars20576Grs6557634 and Grs20575Crs20576Grs6557634 haplotypes conferred two-fold increased risk for GBC. Among these, the DR4 Crs20575Ars20576Ars6557634 haplotype emerged as main factor influencing GBC susceptibility as the risk was not modulated by gender or gallstone stratification. Our meta-analysis results showed significant association of DR4 rs6557634 with overall cancer risk, GI cancers as well as in Caucasians. We didn't find any association of FAS and FASL SNPs with GBC susceptibility. Conclusions The DR4 haplotype Crs20575Ars20576Ars6557634 represents an important factor accounting the patients susceptibility to GBC probably due to decreased apoptosis. However, additional well-designed studies with larger sample size focusing on different ethnicities are required to further validate the results.
Collapse
Affiliation(s)
- Rajani Rai
- Department of Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Kiran L. Sharma
- Department of Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Surbhi Sharma
- Department of Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Sanjeev Misra
- Department of Surgical Oncology, KGMU, Lucknow, India
| | - Ashok Kumar
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Balraj Mittal
- Department of Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
- * E-mail:
| |
Collapse
|
332
|
Lin T, Wan L, Qi X, Shi W, Lin J. A moderate static magnetic field enhances TRAIL-induced apoptosis by the inhibition of Cdc2 and subsequent downregulation of survivin in human breast carcinoma cells. Bioelectromagnetics 2014; 35:337-46. [PMID: 24619849 DOI: 10.1002/bem.21849] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 01/28/2014] [Indexed: 01/04/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exhibits its potent antitumor activity via membrane receptors on cancer cells without deleterious side effects for normal tissue. However, as many other cancer types, breast cancer cells develop a resistance to TRAIL. In the present study, we reported that exposure to 3.0 mT static magnetic field (SMF) mediated the sensitization of breast cancer cells to TRAIL-induced apoptosis. This effect was significantly reduced by the forced expression of survivin, suggesting the sensitization was mediated at least in part through the inhibition of survivin expression. In addition, SMF alone or in combination with TRAIL induced a cell cycle arrest within the G2 /M phase, and the reduction in the survivin protein level was associated with the downregulated expression of Cdc2, a cyclin B-dependent kinase that is necessary for the entry into the M phase. Taken together, our results demonstrated that SMF promoted TRAIL-induced apoptosis by inhibiting the expression of Cdc2 and, subsequently, survivin. Of note, SMF did not sensitize untransformed human mammary epithelial cells to TRAIL-mediated apoptosis. Therefore, the combined treatment of SMF and TRAIL may offer an attractive strategy for safely treating resistant breast cancers.
Collapse
Affiliation(s)
- Tingting Lin
- College of Instrumentation and Electrical Engineering, Jilin University, Changchun, People's Republic of China
| | | | | | | | | |
Collapse
|
333
|
Sun S, Li Z, Sun L, Yang C, Mei Z, Ouyang W, Yang B, Xie C. Results on efficacy and safety of cancer treatment with or without tumor necrosis factor-related apoptosis-inducing ligand-related agents: A meta-analysis. Mol Clin Oncol 2014; 2:440-448. [PMID: 24772315 DOI: 10.3892/mco.2014.261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/21/2014] [Indexed: 11/05/2022] Open
Abstract
This meta-analysis aimed to evaluate the currently available evidence on the efficacy and safety of cancer treatment with or without tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-related agents. We conducted a systematic search through Medline, Cochrane Library and EMBASE electronic databases and manually searched the Journal of Clinical Oncology to identify randomized controlled trials (RCTs) conducted between 1995 and 2013 comparing the efficacy and safety results of cancer treatment with and without TRAIL-related agents. The methodological quality of the included RCTs was assessed by the Cochrane Risk of Bias assessment tool. The outcome measurements included objective response rate (ORR), clinical benefit rate (CBR)/disease control rate (DCR) and adverse events (AEs). The relevant data were analyzed using Review Manager 5.2 software. Grading of Recommendations Assessment Development and Evaluation was used to assess the quality of evidence and grade of recommendation. Four RCTs, including a total of 596 patients, were ultimately selected and analyzed. There were no statistically significant differences among the 4 RCTs regarding ORR [relative risk (RR)=0.92, 95% confidence interval (CI): 0.73-1.15, P=0.45], CBR/DCR (RR=0.92, 95% CI: 0.81-1.05, P=0.21), progression-free survival [hazard ratio (HR)=0.89, 95% CI: 0.75-1.05, P=0.16], overall survival (HR=0.90, 95% CI: 0.74-1.09, P=0.27), number of patients with any AEs (RR=0.99, 95% CI: 0.96-1.03, P=0.77), number of patients with any severe AEs (RR=0.95, 95% CI: 0.78-1.55, P=0.58), number of patients with ≥grade 3 AEs (RR=1.13, 95% CI: 0.93-1.38, P=0.22) and number of fatal AEs (RR=1.14, 95% CI: 0.71-1.81, P=0.59). The quality of evidence was considered to be moderate and the recommendation grades were weak. In conclusion, the benefits of TRAIL-related agents in the treatment of cancer patients remain uncertain and further clinical trials are required.
Collapse
Affiliation(s)
- Shaoxing Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zonghuan Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Li Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Chunxu Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zijie Mei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wen Ouyang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Bo Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China ; ; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
334
|
Novoyatleva T, Sajjad A, Engel FB. TWEAK-Fn14 Cytokine-Receptor Axis: A New Player of Myocardial Remodeling and Cardiac Failure. Front Immunol 2014; 5:50. [PMID: 24611063 PMCID: PMC3920183 DOI: 10.3389/fimmu.2014.00050] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/28/2014] [Indexed: 01/01/2023] Open
Abstract
Tumor necrosis factor (TNF) has been firmly established as a pathogenic factor in heart failure, a significant socio-economic burden. In this review, we will explore the role of other members of the TNF/TNF receptor superfamily (TNFSF/TNFRSF) in cardiovascular diseases (CVDs) focusing on TWEAK and its receptor Fn14, new players in myocardial remodeling and heart failure. The TWEAK/Fn14 pathway controls a variety of cellular activities such as proliferation, differentiation, and apoptosis and has diverse biological functions in pathological mechanisms like inflammation and fibrosis that are associated with CVDs. Furthermore, it has recently been shown that the TWEAK/Fn14 axis is a positive regulator of cardiac hypertrophy and that deletion of Fn14 receptor protects from right heart fibrosis and dysfunction. We discuss the potential use of the TWEAK/Fn14 axis as biomarker for CVDs as well as therapeutic target for future treatment of human heart failure based on supporting data from animal models and in vitro studies. Collectively, existing data strongly suggest the TWEAK/Fn14 axis as a potential new therapeutic target for achieving cardiac protection in patients with CVDs.
Collapse
Affiliation(s)
- Tatyana Novoyatleva
- Department of Cardiac Development and Remodelling, Max-Planck-Institute for Heart and Lung Research , Bad Nauheim , Germany
| | - Amna Sajjad
- Department of Cardiac Development and Remodelling, Max-Planck-Institute for Heart and Lung Research , Bad Nauheim , Germany ; Government College University Faisalabad , Faisalabad , Pakistan
| | - Felix B Engel
- Department of Nephropathology, Experimental Renal and Cardiovascular Research, Institute of Pathology, University of Erlangen-Nürnberg , Erlangen , Germany
| |
Collapse
|
335
|
Kichev A, Rousset CI, Baburamani AA, Levison SW, Wood TL, Gressens P, Thornton C, Hagberg H. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) signaling and cell death in the immature central nervous system after hypoxia-ischemia and inflammation. J Biol Chem 2014; 289:9430-9. [PMID: 24509861 PMCID: PMC3979382 DOI: 10.1074/jbc.m113.512350] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family. The interaction of TRAIL with death receptor 4 (DR4) and DR5 can trigger apoptotic cell death. The aim of this study was to investigate the role of TRAIL signaling in neonatal hypoxia-ischemia (HI). Using a neonatal mouse model of HI, mRNA, and protein expression of TRAIL, DR5 and the TRAIL decoy receptors osteoprotegerin (OPG), mDcTRAILR1, and mDcTRAILR2 were determined. In vitro, mRNA expression of these genes was measured in primary neurons and oligodendrocyte progenitor cells (OPCs) after inflammatory cytokine (TNF-α/IFN-γ) treatment and/or oxygen and glucose deprivation (OGD). The toxicity of these various paradigms was also measured. The expression of TRAIL, DR5, OPG, and mDcTRAILR2 was significantly increased after HI. In vitro, inflammatory cytokines and OGD treatment significantly induced mRNAs for TRAIL, DR5, OPG, and mDcTRAILR2 in primary neurons and of TRAIL and OPG in OPCs. TRAIL protein was expressed primarily in microglia and astroglia, whereas DR5 co-localized with neurons and OPCs in vivo. OGD enhanced TNF-α/IFN-γ toxicity in both neuronal and OPC cultures. Recombinant TRAIL exerted toxicity alone or in combination with OGD and TNF-α/IFN-γ in primary neurons but not in OPC cultures. The marked increases in the expression of TRAIL and its receptors after cytokine exposure and OGD in primary neurons and OPCs were similar to those found in our animal model of neonatal HI. The toxicity of TRAIL in primary neurons suggests that TRAIL signaling participates in neonatal brain injury after inflammation and HI.
Collapse
Affiliation(s)
- Anton Kichev
- From the Centre for the Developing Brain, Perinatal Brain Injury Group, Kings College London, London SE1 7EH, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
336
|
Association of four polymorphisms in the death receptor 4 gene with cancer risk: an updated meta-analysis. Tumour Biol 2014; 35:5149-58. [DOI: 10.1007/s13277-014-1667-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 01/17/2014] [Indexed: 01/19/2023] Open
|
337
|
Abstract
Apoptosis is a cell death program that is well-orchestrated for normal tissue homeostasis and for removal of damaged, old or infected cells. It is regulated by intrinsic and extrinsic pathways. The intrinsic pathway responds to signals such as ultraviolet radiation or DNA damage and activates "executioner" caspases through a mitochondria-dependent pathway. The extrinsic pathway is activated by death signals induced, for example, by an infection that activates the immune system or receptor-mediated pathways. The extrinsic pathway signals also cascade down to executioner caspases that cleave target proteins and lead to cell death. Strict control of cellular apoptosis is important for the hematopoietic system as it has a high turnover rate. However, the apoptosis program is often deregulated in hematologic malignancies leading to the accumulation of malignant cells. Therefore, apoptosis pathways have been identified for the development of anticancer therapeutics. We review here the proteins that have been targeted for anticancer drug development in hematologic malignancies. These include BCL-2 family proteins, death ligands and receptors, inhibitor of apoptosis family proteins and caspases. Except for caspase activators, drugs that target each of these classes of proteins have advanced into clinical trials.
Collapse
Affiliation(s)
- Shadia Zaman
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center , Houston, TX , USA
| | | | | |
Collapse
|
338
|
Cantarella G, Di Benedetto G, Ribatti D, Saccani-Jotti G, Bernardini R. Involvement of caspase 8 and c-FLIPL in the proangiogenic effects of the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL). FEBS J 2014; 281:1505-1513. [PMID: 24438025 DOI: 10.1111/febs.12720] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 12/02/2013] [Accepted: 01/12/2014] [Indexed: 12/26/2022]
Abstract
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), a cytokine of the tumour necrosis factor superfamily, is a potent cell-apoptosis inducer, although its effects vary as a function of concentration. In fact, low concentrations of TRAIL are associated with non-apoptotic effects, such as cell proliferation. Here, the effects of TRAIL at different concentrations have been evaluated on mitogenesis and migration on human umbilical vein endothelial cells (HUVEC) in vitro, as well as in the chick embryo chorioallantoic membrane (CAM) angiogenesis model in vivo. At low concentrations, TRAIL promoted either mitogenesis or migration of HUVEC, evaluated using the wound healing method. Cleavage of caspase 8 was evaluated along with expression of the caspase 8-like molecule, cellular FLICE-inhibitory protein (long form) (c-FLIPL ). Low concentrations of TRAIL failed to induce caspase 8 processing, whereas high concentrations induced apoptosis of HUVEC and activation of caspase 8. Moreover, TRAIL induced a significant angiogenic response in the CAM assay in vivo, comparable with that of vascular endothelial growth factor. These data suggest that the non-apoptotic effects of TRAIL include mitogenesis and increased mobility of endothelial cells, and eventually angiogenesis. In addition, the results demonstrate that the c-FLIPL level is also modulated by differences in TRAIL concentration, suggesting its involvement in the divergent effects of TRAIL. In conclusion, this study envisions a proangiogenic role of TRAIL, suggesting that TRAIL may represent a target for pharmacological manipulation.
Collapse
Affiliation(s)
- Giuseppina Cantarella
- Department of Clinical and Molecular Biomedicine, University of Catania Medical School, Italy
| | | | | | | | | |
Collapse
|
339
|
Brost S, Zimmermann A, Koschny R, Sykora J, Stremmel W, Schirmacher P, Walczak H, Ganten TM. Hepatocyte expression of TRAIL pathway regulators correlates with histopathological and clinical parameters in chronic HCV infection. Pathol Res Pract 2014; 210:83-91. [DOI: 10.1016/j.prp.2013.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/29/2013] [Indexed: 12/15/2022]
|
340
|
Pavet V, Shlyakhtina Y, He T, Ceschin DG, Kohonen P, Perälä M, Kallioniemi O, Gronemeyer H. Plasminogen activator urokinase expression reveals TRAIL responsiveness and supports fractional survival of cancer cells. Cell Death Dis 2014; 5:e1043. [PMID: 24481457 PMCID: PMC4040674 DOI: 10.1038/cddis.2014.5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 11/19/2013] [Accepted: 11/26/2013] [Indexed: 12/24/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/TNFSF10/Apo2L) holds promise for cancer therapy as it induces apoptosis in a large variety of cancer cells while exerting negligible toxicity in normal ones. However, TRAIL can also induce proliferative and migratory signaling in cancer cells resistant to apoptosis induced by this cytokine. In that regard, the molecular mechanisms underlying the tumor selectivity of TRAIL and those balancing apoptosis versus survival remain largely elusive. We show here that high mRNA levels of PLAU, which encodes urokinase plasminogen activator (uPA), are characteristic of cancer cells with functional TRAIL signaling. Notably, decreasing uPA levels sensitized cancer cells to TRAIL, leading to markedly increased apoptosis. Mechanistic analyses revealed three molecular events taking place in uPA-depleted cells: reduced basal ERK1/2 prosurvival signaling, decreased preligand decoy receptor 2 (DcR2)-death receptor 5 (DR5) interaction and attenuated recruitment of DcR2 to the death-inducing signaling complex upon TRAIL challenge. These phenomena were accompanied by increased FADD and procaspase-8 recruitment and processing, thus guiding cells toward a caspase-dependent cell death that is largely independent of the intrinsic apoptosis pathway. Collectively, our results unveil PLAU mRNA levels as marker for the identification of TRAIL-responsive tumor cells and highlight a key role of uPA signaling in ‘apoptosis versus survival' decision-making processes upon TRAIL challenge.
Collapse
Affiliation(s)
- V Pavet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UdS/CERBM, BP10142, 67404 Illkirch Graffenstaden, France
| | - Y Shlyakhtina
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UdS/CERBM, BP10142, 67404 Illkirch Graffenstaden, France
| | - T He
- Medical Biotechnology, VTT Technical Research Centre of Finland and University of Turku, Turku, Finland
| | - D G Ceschin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UdS/CERBM, BP10142, 67404 Illkirch Graffenstaden, France
| | - P Kohonen
- Medical Biotechnology, VTT Technical Research Centre of Finland and University of Turku, Turku, Finland
| | - M Perälä
- Medical Biotechnology, VTT Technical Research Centre of Finland and University of Turku, Turku, Finland
| | - O Kallioniemi
- FIMM-Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - H Gronemeyer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UdS/CERBM, BP10142, 67404 Illkirch Graffenstaden, France
| |
Collapse
|
341
|
Allen JE, Krigsfeld G, Mayes PA, Patel L, Dicker DT, Patel AS, Dolloff NG, Messaris E, Scata KA, Wang W, Zhou JY, Wu GS, El-Deiry WS. Dual inactivation of Akt and ERK by TIC10 signals Foxo3a nuclear translocation, TRAIL gene induction, and potent antitumor effects. Sci Transl Med 2014; 5:171ra17. [PMID: 23390247 DOI: 10.1126/scitranslmed.3004828] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recombinant tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an antitumor protein that is in clinical trials as a potential anticancer therapy but suffers from drug properties that may limit efficacy such as short serum half-life, stability, cost, and biodistribution, particularly with respect to the brain. To overcome such limitations, we identified TRAIL-inducing compound 10 (TIC10), a potent, orally active, and stable small molecule that transcriptionally induces TRAIL in a p53-independent manner and crosses the blood-brain barrier. TIC10 induces a sustained up-regulation of TRAIL in tumors and normal cells that may contribute to the demonstrable antitumor activity of TIC10. TIC10 inactivates kinases Akt and extracellular signal-regulated kinase (ERK), leading to the translocation of Foxo3a into the nucleus, where it binds to the TRAIL promoter to up-regulate gene transcription. TIC10 is an efficacious antitumor therapeutic agent that acts on tumor cells and their microenvironment to enhance the concentrations of the endogenous tumor suppressor TRAIL.
Collapse
Affiliation(s)
- Joshua E Allen
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medicine-Hematology/Oncology, Penn State Hershey Cancer Institute, Hershey, PA 17033, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
342
|
Selective CDK9 inhibition overcomes TRAIL resistance by concomitant suppression of cFlip and Mcl-1. Cell Death Differ 2013; 21:491-502. [PMID: 24362439 PMCID: PMC3921597 DOI: 10.1038/cdd.2013.179] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 10/07/2013] [Accepted: 11/05/2013] [Indexed: 02/04/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can induce apoptosis in many cancer cells without causing toxicity in vivo. However, to date, TRAIL-receptor agonists have only shown limited therapeutic benefit in clinical trials. This can, most likely, be attributed to the fact that 50% of all cancer cell lines and most primary human cancers are TRAIL resistant. Consequently, future TRAIL-based therapies will require the addition of sensitizing agents that remove crucial blocks in the TRAIL apoptosis pathway. Here, we identify PIK-75, a small molecule inhibitor of the p110α isoform of phosphoinositide-3 kinase (PI3K), as an exceptionally potent TRAIL apoptosis sensitizer. Surprisingly, PI3K inhibition was not responsible for this activity. A kinome-wide in vitro screen revealed that PIK-75 strongly inhibits a panel of 27 kinases in addition to p110α. Within this panel, we identified cyclin-dependent kinase 9 (CDK9) as responsible for TRAIL resistance of cancer cells. Combination of CDK9 inhibition with TRAIL effectively induced apoptosis even in highly TRAIL-resistant cancer cells. Mechanistically, CDK9 inhibition resulted in downregulation of cellular FLICE-like inhibitory protein (cFlip) and Mcl-1 at both the mRNA and protein levels. Concomitant cFlip and Mcl-1 downregulation was required and sufficient for TRAIL sensitization by CDK9 inhibition. When evaluating cancer selectivity of TRAIL combined with SNS-032, the most selective and clinically used inhibitor of CDK9, we found that a panel of mostly TRAIL-resistant non-small cell lung cancer cell lines was readily killed, even at low concentrations of TRAIL. Primary human hepatocytes did not succumb to the same treatment regime, defining a therapeutic window. Importantly, TRAIL in combination with SNS-032 eradicated established, orthotopic lung cancer xenografts in vivo. Based on the high potency of CDK9 inhibition as a cancer cell-selective TRAIL-sensitizing strategy, we envisage the development of new, highly effective cancer therapies.
Collapse
|
343
|
Qiu F, Hu M, Tang B, Liu X, Zhuang H, Yang J, Hua ZC. Annexin V-TRAIL fusion protein is a more sensitive and potent apoptotic inducer for cancer therapy. Sci Rep 2013; 3:3565. [PMID: 24356445 PMCID: PMC3868960 DOI: 10.1038/srep03565] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 12/04/2013] [Indexed: 11/17/2022] Open
Abstract
The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising cancer therapeutic agent, which kills cancer cells selectively, while leaving normal cells unharmed. However, the emerging resistance of tumor cells and patients to TRAIL-induced apoptosis limits its further application. In this study, we developed a chimeric protein Annexin V-TRAIL (designated as TP8) with higher efficacy than TRAIL both in vitro and in vivo. In vitro, the EC50 of TP8 on a series of tumor cells was much lower than wild-type TRAIL. Annexin V provided this recombinant protein with higher efficacy, while leaving tumor specificity of TRAIL unchanged since TP8 had no effects on normal cells. Invivo, TP8 effectively suppressed tumor growth and prolonged tumor doubling time and tumor growth delay time in mouse xenografts involving multiple cancer cell types including A549, Colo205 and Bel7402. This study provides a new rational strategy to treat TRAIL-resistant cancers.
Collapse
Affiliation(s)
- Fan Qiu
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences and School of Stomatology, Affiliated Stomatological Hospital, Nanjing University, Nanjing, 210093, P.R. China
| | - Minjin Hu
- Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc., Changzhou, 213164, P.R. China
| | - Bo Tang
- Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc., Changzhou, 213164, P.R. China
| | - Xiufeng Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences and School of Stomatology, Affiliated Stomatological Hospital, Nanjing University, Nanjing, 210093, P.R. China
| | - Hongqin Zhuang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences and School of Stomatology, Affiliated Stomatological Hospital, Nanjing University, Nanjing, 210093, P.R. China
| | - Jie Yang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences and School of Stomatology, Affiliated Stomatological Hospital, Nanjing University, Nanjing, 210093, P.R. China
| | - Zi-Chun Hua
- 1] The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences and School of Stomatology, Affiliated Stomatological Hospital, Nanjing University, Nanjing, 210093, P.R. China [2] Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc., Changzhou, 213164, P.R. China
| |
Collapse
|
344
|
|
345
|
Marriott I. Apoptosis-associated uncoupling of bone formation and resorption in osteomyelitis. Front Cell Infect Microbiol 2013; 3:101. [PMID: 24392356 PMCID: PMC3867676 DOI: 10.3389/fcimb.2013.00101] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 12/04/2013] [Indexed: 01/18/2023] Open
Abstract
The mechanisms underlying the destruction of bone tissue in osteomyelitis are only now being elucidated. While some of the tissue damage associated with osteomyelitis likely results from the direct actions of bacteria and infiltrating leukocytes, perhaps exacerbated by bacterial manipulation of leukocyte survival pathways, infection-induced bone loss predominantly results from an uncoupling of the activities of osteoblasts and osteoclasts. Bacteria or their products can directly increase osteoclast formation and activity, and the inflammatory milieu at sites of infection can further promote bone resorption. In addition, osteoclast activity is critically regulated by osteoblasts that can respond to bacterial pathogens and foster both inflammation and osteoclastogenesis. Importantly, bone loss during osteomyelitis is also brought about by a decline in new bone deposition due to decreased bone matrix synthesis and by increased rates of osteoblast apoptosis. Extracellular bacterial components may be sufficient to reduce osteoblast viability, but the causative agents of osteomyelitis are also capable of inducing continuous apoptosis of these cells by activating intrinsic and extrinsic cell death pathways to further uncouple bone formation and resorption. Interestingly, bacterial internalization appears to be required for maximal osteoblast apoptosis, and cytosolic inflammasome activation may act in concert with autocrine/paracrine death receptor-ligand signaling to induce cell death. The manipulation of apoptotic pathways in infected bone cells could be an attractive new means to limit inflammatory damage in osteomyelitis. However, the mechanism that is the most important in bacterium-induced bone loss has not yet been identified. Furthermore, it remains to be determined whether the host would be best served by preventing osteoblast cell death or by promoting apoptosis in infected cells.
Collapse
Affiliation(s)
- Ian Marriott
- Department of Biology, University of North Carolina at Charlotte Charlotte, NC, USA
| |
Collapse
|
346
|
Huo W, Jin N, Fan L, Wang W. MiRNA regulation of TRAIL expression exerts selective cytotoxicity to prostate carcinoma cells. Mol Cell Biochem 2013; 388:123-33. [PMID: 24292881 DOI: 10.1007/s11010-013-1904-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 11/15/2013] [Indexed: 12/17/2022]
Abstract
Prostate carcinoma is the most common cancer for men and among the leading cancer-related causes. Many evidences have shown that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) potently induces apoptosis in cancer cells, and thus, is a promising biologic agent for prostate carcinoma therapy. However, TRAIL expression mediated by the current vectors lacks tumor specificity, thereby exerting cytotoxicity to normal cells. To solve this problem, we inserted miRNA response elements (MREs), miR-143 and miR-145, expression levels of which were reduced in prostate carcinoma, as well as that of miR-122, which is specifically expressed in hepatic cells, into adenoviral vectors to control TRAIL expression (Ad-TRAIL-M3). qPCR data confirmed that miR-143, miR-145, and miR-122 levels were all decreased in prostate carcinoma cell lines and prostate cancer samples from patients. Luciferase assays showed that MREs-regulated luciferase expression was potently suppressed in normal cells, but not in prostate cancer cells. Ad-TRAIL-M3, which expresses TRAIL in a MREs-regulated manner, produced high level of TRAIL and suppressed the survival of prostate cancer cells by inducing apoptosis, while Ad-TRAIL-M3 had no TRAIL expression in normal cells and thus exerted no cytotoxicity to them. The studies on PC-3 tumor xenograft in mice further confirmed that Ad-TRAIL-M3 was able to inhibit the growth of tumors and possessed high biosafety. In conclusion, we successfully generated an adenoviral vector that expresses TRAIL in miRNA-regulated mechanism. This miRNA-based gene therapy may be promising for prostate carcinoma treatment.
Collapse
Affiliation(s)
- Wei Huo
- Department of Urology, China Japan Union Hospital of Jilin University, Changchun, 130033, China
| | | | | | | |
Collapse
|
347
|
Tatsuta T, Hosono M, Takahashi K, Omoto T, Kariya Y, Sugawara S, Hakomori S, Nitta K. Sialic acid-binding lectin (leczyme) induces apoptosis to malignant mesothelioma and exerts synergistic antitumor effects with TRAIL. Int J Oncol 2013; 44:377-84. [PMID: 24297392 PMCID: PMC3898873 DOI: 10.3892/ijo.2013.2192] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/17/2013] [Indexed: 11/06/2022] Open
Abstract
Malignant mesothelioma is a highly aggressive tumor with poor prognosis. An effective drug for treatment of malignant mesothelioma is greatly needed. Sialic acid-binding lectin (SBL) isolated from oocytes of Rana catesbeiana is a multifunctional protein which has lectin activity, ribonuclease activity and antitumor activity, so it could be developed as a new type of anticancer drug. The validity of SBL for treatment of malignant mesothelioma was assessed using three malignant mesotheliomas and a non-malignant mesothlial cell line. Effectiveness of combinatorial treatment of SBL and tumor necrosis factor-related apoptosis inducing ligand (TRAIL) was also elucidated and characterized. SBL induced tumor-selective cytotoxicity that was attributed to induction of apoptosis. Combinatorial treatment of SBL and TRAIL showed synergistic apoptosis-inducing effect. Additional experiments revealed that Bid was the mediating molecule for the synergistic effect in SBL and TRAIL. These results suggested that SBL could be a promising candidate for the therapeutics for malignant mesothelioma. Furthermore, the combinatorial treatment of SBL and TRAIL could be an effective regimen against malignant mesothelioma.
Collapse
Affiliation(s)
- Takeo Tatsuta
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai 981-8558, Japan
| | - Masahiro Hosono
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai 981-8558, Japan
| | - Kohta Takahashi
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai 981-8558, Japan
| | - Takashi Omoto
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai 981-8558, Japan
| | - Yukiko Kariya
- Fukushima Medical University, Fukushima 960-1295, Japan
| | - Shigeki Sugawara
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai 981-8558, Japan
| | - Senitiroh Hakomori
- Division of Biomembrane Research, Pacific Northwest Research Institute, WA 98122, USA
| | - Kazuo Nitta
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai 981-8558, Japan
| |
Collapse
|
348
|
Koyama M, Sowa Y, Horinaka M, Goda AE, Fujiwara J, Sakai T. Peroxisome proliferator-activated receptor γ ligand troglitazone and TRAIL synergistically induce apoptosis. Oncol Rep 2013; 31:947-54. [PMID: 24276615 DOI: 10.3892/or.2013.2868] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 10/25/2013] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is known to cause apoptosis in several types of malignant tumor cells through its interaction with the death domain-containing receptor, death receptor 5 (DR5). In the present study, we showed that co-treatment with troglitazone (TGZ), a synthetic ligand of peroxisome proliferator-activated receptor γ (PPARγ), and TRAIL synergistically induced apoptosis through DR5 upregulation in human colon cancer DLD-1 cells. TGZ elevated DR5 expression at the promoter level through the CCAAT/enhancer-binding protein homologous protein (CHOP) binding site. These results suggest that combined treatment with TGZ and TRAIL may be promising as a new therapy against malignant tumors.
Collapse
Affiliation(s)
- Makoto Koyama
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yoshihiro Sowa
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Mano Horinaka
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Ahmed E Goda
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Jun Fujiwara
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Toshiyuki Sakai
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
349
|
Wang C, Xu S, Yi F, Wang X, Lei Y, Huang S, Zhou R, Xia B. Tumor Necrosis Factor-Related Apoptosis Inducing Ligand Gene Polymorphisms are Correlated with Gastric Cancer in Central China. Pharm Res 2013; 32:762-8. [DOI: 10.1007/s11095-013-1217-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 09/20/2013] [Indexed: 02/07/2023]
|
350
|
Ethanolic Extract of Polish Propolis: Chemical Composition and TRAIL-R2 Death Receptor Targeting Apoptotic Activity against Prostate Cancer Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:757628. [PMID: 24324518 PMCID: PMC3845518 DOI: 10.1155/2013/757628] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 09/18/2013] [Indexed: 12/03/2022]
Abstract
Propolis possesses chemopreventive properties through direct anticancer and indirect immunomodulatory activities. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) plays a significant role in immunosurveillance and defense against cancer cells. TRAIL triggers apoptosis upon binding to TRAIL-R1 (DR4) and TRAIL-R2 (DR5) death receptors expressed on cancer cell surface. The activation of TRAIL apoptotic signaling is considered an attractive option for cancer prevention. However, as more tumor cells are reported to be resistant to TRAIL-mediated death, it is important to develop new strategies to overcome this resistance. The aim of this study was to investigate the chemical composition and proapoptotic mechanism of ethanolic extract of Polish propolis (EEP-P) against cancer cells. The identification and quantification of phenolic compounds in propolis extract were performed using HPLC-DAD and UPLC-Q-TOF-MS methods. TRAIL-resistant LNCaP prostate cancer cells were treated with EEP-P and TRAIL. Cytotoxicity was measured by MTT and LDH assays. Apoptosis was detected using annexin V-FITC staining by flow cytometry and fluorescence microscopy. Death receptors expression was analyzed using flow cytometry. Pinobanksin, chrysin, methoxyflavanone, p-coumaric acid, ferulic acid and caffeic acid were the main phenolics found in EEP-P. Propolis sensitized LNCaP cells through upregulation of TRAIL-R2. These results suggest that EEP-P supports TRAIL-mediated immunochemoprevention in prostate cancer cells.
Collapse
|