1
|
Tuomela K, Ambrose AR, Davis DM. Escaping Death: How Cancer Cells and Infected Cells Resist Cell-Mediated Cytotoxicity. Front Immunol 2022; 13:867098. [PMID: 35401556 PMCID: PMC8984481 DOI: 10.3389/fimmu.2022.867098] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/04/2022] [Indexed: 12/14/2022] Open
Abstract
Cytotoxic lymphocytes are critical in our immune defence against cancer and infection. Cytotoxic T lymphocytes and Natural Killer cells can directly lyse malignant or infected cells in at least two ways: granule-mediated cytotoxicity, involving perforin and granzyme B, or death receptor-mediated cytotoxicity, involving the death receptor ligands, tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and Fas ligand (FasL). In either case, a multi-step pathway is triggered to facilitate lysis, relying on active pro-death processes and signalling within the target cell. Because of this reliance on an active response from the target cell, each mechanism of cell-mediated killing can be manipulated by malignant and infected cells to evade cytolytic death. Here, we review the mechanisms of cell-mediated cytotoxicity and examine how cells may evade these cytolytic processes. This includes resistance to perforin through degradation or reduced pore formation, resistance to granzyme B through inhibition or autophagy, and resistance to death receptors through inhibition of downstream signalling or changes in protein expression. We also consider the importance of tumour necrosis factor (TNF)-induced cytotoxicity and resistance mechanisms against this pathway. Altogether, it is clear that target cells are not passive bystanders to cell-mediated cytotoxicity and resistance mechanisms can significantly constrain immune cell-mediated killing. Understanding these processes of immune evasion may lead to novel ideas for medical intervention.
Collapse
Affiliation(s)
| | | | - Daniel M. Davis
- The Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
2
|
Lim B, Greer Y, Lipkowitz S, Takebe N. Novel Apoptosis-Inducing Agents for the Treatment of Cancer, a New Arsenal in the Toolbox. Cancers (Basel) 2019; 11:cancers11081087. [PMID: 31370269 PMCID: PMC6721450 DOI: 10.3390/cancers11081087] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/11/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023] Open
Abstract
Evasion from apoptosis is an important hallmark of cancer cells. Alterations of apoptosis pathways are especially critical as they confer resistance to conventional anti-cancer therapeutics, e.g., chemotherapy, radiotherapy, and targeted therapeutics. Thus, successful induction of apoptosis using novel therapeutics may be a key strategy for preventing recurrence and metastasis. Inhibitors of anti-apoptotic molecules and enhancers of pro-apoptotic molecules are being actively developed for hematologic malignancies and solid tumors in particular over the last decade. However, due to the complicated apoptosis process caused by a multifaceted connection with cross-talk pathways, protein–protein interaction, and diverse resistance mechanisms, drug development within the category has been extremely challenging. Careful design and development of clinical trials incorporating predictive biomarkers along with novel apoptosis-inducing agents based on rational combination strategies are needed to ensure the successful development of these molecules. Here, we review the landscape of currently available direct apoptosis-targeting agents in clinical development for cancer treatment and update the related biomarker advancement to detect and validate the efficacy of apoptosis-targeted therapies, along with strategies to combine them with other agents.
Collapse
Affiliation(s)
- Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Yoshimi Greer
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Stanley Lipkowitz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Naoko Takebe
- Early Clinical Trials Development, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
3
|
Relationship between the Regulation of Caspase-8-Mediated Apoptosis and Radioresistance in Human THP-1-Derived Macrophages. Int J Mol Sci 2018; 19:ijms19103154. [PMID: 30322167 PMCID: PMC6214119 DOI: 10.3390/ijms19103154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/03/2018] [Accepted: 10/12/2018] [Indexed: 12/24/2022] Open
Abstract
Radiosensitivity varies depending on the cell type; highly differentiated cells typically exhibit greater radioresistance. We recently demonstrated that human macrophages derived from THP-1 monocytic cells, which lack TP53, are highly resistant to radiation-induced apoptosis compared with undifferentiated THP-1 cells. However, the mechanisms by which THP-1 cells acquire radioresistance during differentiation remain unknown. Herein, we investigated the mechanisms by which THP-1-derived macrophages develop p53-independent radioresistance by analyzing DNA damage responses and apoptotic pathways. Analysis of γ-H2AX foci, which indicates the formation of DNA double-strand breaks (DSB), suggested that a capacity to repair DSB of macrophages is comparable to that of radiosensitive THP-1 cells. Furthermore, treatment with inhibitors against DSB repair-related proteins failed to enhance radiation-induced apoptosis in THP-1-derrived macrophages. Analysis of the apoptotic pathways showed that radiosensitive THP-1 cells undergo apoptosis through the caspase-8/caspase-3 cascade after irradiation, whereas this was not observed in the macrophages. Caspase-8 protein expression was lower in macrophages than in THP-1 cells, whereas mRNA expressions were comparable between both cell types. Co-treatment with a proteasome inhibitor and ionizing radiation effectively induced apoptosis in macrophages in a caspase-8-dependent manner. Results suggest that the regulation of caspase-8-mediated apoptosis during differentiation plays a role in the p53-independent radioresistance of THP-1-derived macrophages.
Collapse
|
4
|
Sato Y, Yoshino H, Kazama Y, Kashiwakura I. Involvement of caspase‑8 in apoptosis enhancement by cotreatment with retinoic acid‑inducible gene‑I‑like receptor agonist and ionizing radiation in human non‑small cell lung cancer. Mol Med Rep 2018; 18:5286-5294. [PMID: 30320341 DOI: 10.3892/mmr.2018.9536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 09/07/2018] [Indexed: 11/06/2022] Open
Abstract
Retinoic acid‑inducible gene‑I‑like receptors (RLRs) serve an important role in antiviral immune responses. Recent studies demonstrated that RLR activation exerts antitumor activity by inducing an anticancer immune response and apoptosis in various cancer cells. The authors' recent study demonstrated that the cytotoxic effects of the RLR agonist Poly(I:C)‑HMW/LyoVec™ [Poly(I:C)‑HMW] in human non‑small cell lung cancer (NSCLC) were enhanced by cotreatment with ionizing radiation (IR). Furthermore, cotreatment with Poly(I:C)‑HMW and IR effectively induced cell death, including apoptosis, in a caspase‑dependent manner. However, the mechanisms by which cotreatment with Poly(I:C)‑HMW and IR effectively induce apoptosis remains unclear. Therefore, the pathways involved in the increase in apoptosis elicited by cotreatment with Poly(I:C)‑HMW and IR in the A549 human NSCLC cell line were investigated. Poly(I:C)‑HMW induced the expression of active caspase‑8 and ‑9, and the Poly(I:C)‑HMW‑induced increase in the cell cycle sub‑G1 population, which is one of the hallmarks of apoptosis, was decreased by treatment with a caspase‑8 inhibitor and caspase‑9 inhibitor. When cells were treated with Poly(I:C)‑HMW and IR, the sub‑G1 population, and the active caspase‑8 and caspase‑9 expression were all increased compared with cells treated with Poly(I:C)‑HMW or IR alone. Furthermore, expression of X‑linked inhibitor of apoptosis protein, which negatively regulates caspase activation, was decreased in cells cotreated with Poly(I:C)‑HMW and IR. Notably, treatment with an inhibitor for caspase‑8, not caspase‑9, partially reversed the net increase in the sub‑G1 population induced by cotreatment with Poly(I:C)‑HMW and IR. Collectively, these results suggested that Poly(I:C)‑HMW induces apoptosis through caspase‑8 and caspase‑9 activation; however, the apoptotic pathway mediated by casapse‑8, and not casapse‑9, is involved in the enhancement of apoptosis caused by cotreatment with Poly(I:C)‑HMW and IR.
Collapse
Affiliation(s)
- Yoshiaki Sato
- Department of Radiation Science, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036‑8564, Japan
| | - Hironori Yoshino
- Department of Radiation Science, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036‑8564, Japan
| | - Yuka Kazama
- Department of Radiological Technology, Hirosaki University School of Health Sciences, Hirosaki, Aomori 036‑8564, Japan
| | - Ikuo Kashiwakura
- Department of Radiation Science, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036‑8564, Japan
| |
Collapse
|
5
|
Estlack LE, Roth CC, Thompson GL, Lambert WA, Ibey BL. Nanosecond pulsed electric fields modulate the expression of Fas/CD95 death receptor pathway regulators in U937 and Jurkat Cells. Apoptosis 2014; 19:1755-68. [DOI: 10.1007/s10495-014-1041-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
6
|
Ifeadi V, Garnett-Benson C. Sub-lethal irradiation of human colorectal tumor cells imparts enhanced and sustained susceptibility to multiple death receptor signaling pathways. PLoS One 2012; 7:e31762. [PMID: 22389673 PMCID: PMC3289623 DOI: 10.1371/journal.pone.0031762] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 01/18/2012] [Indexed: 01/01/2023] Open
Abstract
Background Death receptors (DR) of the TNF family function as anti-tumor immune effector molecules. Tumor cells, however, often exhibit DR-signaling resistance. Previous studies indicate that radiation can modify gene expression within tumor cells and increase tumor cell sensitivity to immune attack. The aim of this study is to investigate the synergistic effect of sub-lethal doses of ionizing radiation in sensitizing colorectal carcinoma cells to death receptor-mediated apoptosis. Methodology/Principal Findings The ability of radiation to modulate the expression of multiple death receptors (Fas/CD95, TRAILR1/DR4, TRAILR2/DR5, TNF-R1 and LTβR) was examined in colorectal tumor cells. The functional significance of sub-lethal doses of radiation in enhancing tumor cell susceptibility to DR-induced apoptosis was determined by in vitro functional sensitivity assays. The longevity of these changes and the underlying molecular mechanism of irradiation in sensitizing diverse colorectal carcinoma cells to death receptor-mediated apoptosis were also examined. We found that radiation increased surface expression of Fas, DR4 and DR5 but not LTβR or TNF-R1 in these cells. Increased expression of DRs was observed 2 days post-irradiation and remained elevated 7-days post irradiation. Sub-lethal tumor cell irradiation alone exhibited minimal cell death, but effectively sensitized three of three colorectal carcinoma cells to both TRAIL and Fas-induced apoptosis, but not LTβR-induced death. Furthermore, radiation-enhanced Fas and TRAIL-induced cell death lasted as long as 5-days post-irradiation. Specific analysis of intracellular sensitizers to apoptosis indicated that while radiation did reduce Bcl-XL and c-FLIP protein expression, this reduction did not correlate with the radiation-enhanced sensitivity to Fas and/or TRAIL mediated apoptosis among the three cell types. Conclusions/Significance Irradiation of tumor cells can overcome Fas and TRAIL resistance that is long lasting. Overall, results of these investigations suggest that non-lethal doses of radiation can be used to make human tumors more amenable to attack by anti-tumor effector molecules and cells.
Collapse
Affiliation(s)
| | - Charlie Garnett-Benson
- Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
7
|
Radiotherapy and TRAIL for cancer therapy. Cancer Lett 2011; 332:184-93. [PMID: 21824725 DOI: 10.1016/j.canlet.2011.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 06/10/2011] [Accepted: 07/02/2011] [Indexed: 11/22/2022]
Abstract
The use of radiotherapy and concomitant chemotherapy substantially improved cure rates in patients with different malignant tumours. However, it is unlikely that further improvements based on conventional chemotherapy may be achieved in the future since increased rates of acute side effects already limit the value of these approaches. Additionally, the increased local control rates are counterweighted by still high rates of distant failures resulting in low net gains for the patients. Thus, there is a currently unmet need for the integration of target-specific drugs improving local control as well distant control into radiation based treatment protocols. In this regard, the death-receptor ligand TNF-α-related apoptosis-inducing ligand (TRAIL/Apo2L) and TRAIL-receptor agonistic antibodies were shown to display a high selectivity for tumour cells and act synergistically with conventional chemotherapy drugs and radiation. Up to now it has been shown that radiation strongly sensitises malignant cells to TRAIL and TRAIL-agonistic antibodies. Synergistic induction of apoptosis was demonstrated in a majority of malignant cell types and xenograft models. Especially in those cells types displaying only weak responses to either treatment alone, strong sensitising effects were described. Moreover, in merely all normal cells and tissues no synergistic effects were found. Depending on cell type and experimental setting, the efficacy of combined treatment is determined by the p53-status, the balance between pro- and anti-apoptotic Bcl-2 proteins and modulation of TRAIL-receptor signal transduction.
Collapse
|
8
|
In vitro study on apoptosis induced by strontium-89 in human breast carcinoma cell line. J Biomed Biotechnol 2011; 2011:541487. [PMID: 21716903 PMCID: PMC3116624 DOI: 10.1155/2011/541487] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 03/24/2011] [Accepted: 03/25/2011] [Indexed: 11/17/2022] Open
Abstract
Many radiopharmaceuticals used for medical diagnosis and therapy are beta emitters; however, the mechanism of the cell death caused by beta-irradiation is not well understood. The objective of this study was to investigate the apoptosis of human breast carcinoma MCF-7 cell lines induced by Strontium-89 (⁸⁹Sr) and its regulation and control mechanism. High-metastatic Breast Carcinoma MCF-7 cells were cultured in vitro using ⁸⁹Sr with different radioactive concentration. The inhibition rate of cell proliferation was measured by MTT color matching method. The cell cycle retardation, apoptosis conditions, mitochondrion transmembrane potential difference and Fas expression were tested and analyzed. The genes P53 and bcl-2 expressions was also analyzed using immunity histochemical analysis. After being induced by ⁸⁹Sr with various of radioactive concentration, it was found that the inhibition of cell proliferation of MCF-7 cells was obviously, the retardation of cell cycle occurred mainly in G2-M. It was also found that the obvious apoptosis occurred after being induced by ⁸⁹Sr, the highest apoptosis rate reached 46.28%. The expressions of Fas acceptor and P53 gene increased, while bcl-2 gene expression decreasesd. These findings demonstrate that in the ranges of a certain radioactive concentration, the inhibition rate of MCF-7 cell proliferation and retardation of cell cycle had positive correlation with the concentration of ⁸⁹Sr. And the mitochondrion transmembrane potential decrease would induce the apoptosis of MCF-7 cell notably, which were controlled by P53 and bcl-2 genes, involved with the Fas acceptor.
Collapse
|
9
|
Chandrasekharan DK, Khanna PK, Kagiya TV, Nair CKK. Synthesis of Nanosilver Using a Vitamin C Derivative and Studies on Radiation Protection. Cancer Biother Radiopharm 2011; 26:249-57. [DOI: 10.1089/cbr.2010.0862] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
10
|
Meike S, Yamamori T, Yasui H, Eitaki M, Matsuda A, Inanami O. 8-Aminoadenosine enhances radiation-induced cell death in human lung carcinoma A549 cells. JOURNAL OF RADIATION RESEARCH 2011; 52:456-463. [PMID: 21785234 DOI: 10.1269/jrr.10188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The combination of a chemotherapeutic agent and radiation is widely applied to enhance cell death in solid tumor cells in cancer treatment. The purine analogue 8-aminoadenosine (8-NH(2)-Ado) is known to be a transcription inhibitor that has proved very effective in multiple myeloma cell lines and primary indolent leukemia cells. In this report, to examine whether 8-NH(2)-Ado had the ability to enhance the radiation-induced cell killing in solid tumor cells, human lung adenocarcinoma A549 cells were irradiated in the presence and absence of 8-NH(2)-Ado. 8-NH(2)-Ado significantly increased reproductive cell death and apoptosis in A549 cells exposed to X-rays. When peptide inhibitors against caspase-3, -8, and -9 were utilized to evaluate the involvement of caspases, all inhibitors suppressed the enhancement of radiation-induced apoptosis, suggesting that not only mitochondria-mediated apoptotic signal transduction pathways but also death receptor-mediated pathways were involved in this enhancement of apoptosis. In addition, in the cells exposed to the treatment combining X-irradiation and 8-NH(2)-Ado, reduction of the intracellular ATP concentration was essential for survival, and down-regulation of the expression of antiapoptotic proteins such as survivin and XIAP was observed. These results indicate that 8-NH(2)-Ado has potential not only as an anti-tumor drug for leukemia and lymphoma but also as a radiosensitizing agent for solid tumors.
Collapse
Affiliation(s)
- Shunsuke Meike
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Japan
| | | | | | | | | | | |
Collapse
|
11
|
TRAIL-induced apoptosis and expression of death receptor TRAIL-R1 and TRAIL-R2 in bladder cancer cells. Folia Histochem Cytobiol 2010; 47:579-85. [PMID: 20430723 DOI: 10.2478/v10042-009-0111-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo2L) is a member of TNF superfamily able to induce programmed death in cancer cells with no toxicity against normal tissues. TRAIL mediate apoptosis follows binding to the two death receptors, TRAIL-R1 (DR4) and/or TRAIL-R2 (DR5). In this study we investigated the cytotoxic and apoptotic effect of TRAIL on bladder cancer cells and the expression of death receptor TRAIL-R1 and TRAIL-R2 on the surface of these cancer cells. Three human bladder transitional cancer cell (TCC) lines - SW780, 647V and T24 were tested for TRAIL sensitivity. The bladder cancer cells were incubated with human soluble recombinant TRAIL. Cytotoxicity was measured by MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-dimethyltetrazolium bromide) and LDH (lactate dyhydrogenase) assays. Apoptosis was detected by flow cytometry with annexin V-FITC/propidium iodide and by fluorescence microscopy with Hoechst 33342/annexin V-FITC/Ethidium Homodimer. The cell surface expression of TRAIL death receptors on bladder cancer were determined using flow cytometry with phycoerythrin-conjugated monoclonal anti-human TRAIL-R1 and TRAIL-R2. Our investigations confirmed that SW780 cells were sensitive to TRAIL, and two other bladder cancer cell lines, 647V and T24, were resistant to TRAIL induced apoptosis. We therefore examined the expression of TRAIL death receptors on bladder cancer cell surfaces. We showed decreased expression of TRAIL-R2 receptor in TRAIL-resistant bladder cancer cells and increased expression of this death receptor in TRAIL-sensitive SW780 cells. The expression of TRAILR1 receptor was similar in all bladder cancer cell lines. TRAIL is one of the promising candidates for cancer therapeutics. However, some cancer cells are resistant to TRAIL-mediated apoptosis. It is therefore important to overcome this resistance for the clinical use of TRAIL in cancer therapy. TRAIL death receptors are attractive therapeutic targets in cancer treatment. The cytotoxic agents capable of up-regulating the expression of TRAIL-R1 and TRAIL-R2 can sensitize cancer cells to TRAIL induced apoptosis.
Collapse
|
12
|
Hori T, Kondo T, Kanamori M, Tabuchi Y, Ogawa R, Zhao QL, Ahmed K, Yasuda T, Seki S, Suzuki K, Kimura T. Ionizing radiation enhances tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through up-regulations of death receptor 4 (DR4) and death receptor 5 (DR5) in human osteosarcoma cells. J Orthop Res 2010; 28:739-45. [PMID: 20041491 DOI: 10.1002/jor.21056] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Despite improvements in chemotherapy and surgery in the treatment of osteosarcoma (OS), satisfactory results are still difficult to achieve. Novel therapeutic modalities need to be developed for osteosarcoma treatment. The combined effects of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and ionizing radiation (IR) on human OS cells were investigated. IR and TRAIL treatment synergistically decreased the cell viability and enhanced apoptosis in OS cell lines. IR pretreatment enhances TRAIL-induced Bid and caspase-3 activations. Decreases in the expression levels of the antiapoptotic proteins c-FLIP and XIAP also associated with apoptosis enhancement. Furthermore, IR pretreatment enhanced DR4 and DR5 expressions at the transcription stage. These results can become the basic lines of evidence for the future treatment of OS using TRAIL with IR.
Collapse
Affiliation(s)
- Takeshi Hori
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Science, University of Toyama, Toyama-city, Toyama, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Koka PS, Mondal D, Schultz M, Abdel-Mageed AB, Agrawal KC. Studies on molecular mechanisms of growth inhibitory effects of thymoquinone against prostate cancer cells: role of reactive oxygen species. Exp Biol Med (Maywood) 2010; 235:751-60. [DOI: 10.1258/ebm.2010.009369] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Thymoquinone (TQ), an active ingredient of black seed oil ( Nigella Sativa), has been shown to possess antineoplastic activity against a variety of experimental tumors. However, the precise mechanism of action of TQ is not known. We investigated the mechanism of action of TQ in androgen receptor (AR)-independent (C4-2B) and AR naïve (PC-3) prostate cancer cells, as models of aggressive prostate cancers. Exposure (24–48 h) to TQ (25–150 μmol/L) inhibited the growth of both C4-2B and PC-3 cells, with IC50 values of approximately 50 and 80 μmol/L, respectively. Within one hour, TQ increased reactive oxygen species (ROS) levels (3-fold) and decreased glutathione (GSH) levels (60%) in both cell types. Pretreatment with N-acetylcysteine (NAC) inhibited both TQ-induced ROS generation and growth inhibition. TQ did not increase the activity of caspases and the caspase inhibitor, z-VAD-FMK did not decrease TQ-induced apoptosis. Furthermore, although TQ treatment resulted in the activation of Jun kinase (JNK), pretreatment with the JNK inhibitor, SP600125, did not protect cells from TQ. However, TQ significantly up-regulated the expressions of growth arrest and DNA damage inducible gene (GADD45 α) and apoptosis-inducing factor-1 and down-regulated the expressions of several Bc12-related proteins, such as BAG-1, Bcl2, Bcl2A1, Bcl2L1 and BID. In C4-2B cells, TQ dose dependently inhibited both total and nuclear AR levels (4–5 fold) and AR-directed transcriptional activity (10–12 fold). Interestingly, this suppressive effect on AR was not prevented by NAC, which clearly suggested that TQ-induced cytotoxicity is not due to changes in AR regulation. These data suggest that TQ-induced cell death is primarily due to increased ROS generation and decreased GSH levels, and is independent of AR activity.
Collapse
Affiliation(s)
| | | | | | - Asim B Abdel-Mageed
- Department of Urology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | |
Collapse
|
14
|
Ermakov AV, Konkova MS, Kostyuk SV, Egolina NA, Efremova LV, Veiko NN. Oxidative stress as a significant factor for development of an adaptive response in irradiated and nonirradiated human lymphocytes after inducing the bystander effect by low-dose X-radiation. Mutat Res 2009; 669:155-161. [PMID: 19540246 DOI: 10.1016/j.mrfmmm.2009.06.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2008] [Revised: 06/04/2009] [Accepted: 06/11/2009] [Indexed: 05/27/2023]
Abstract
X-radiation (10cGy) was shown to induce in human lymphocytes transposition of homologous chromosomes loci from the membrane towards the centre of the nucleus and activation of the chromosomal nucleolus-forming regions (NFRs). These effects are transmitted by means of extracellular DNA (ecDNA) fragments to nonirradiated cells (the so-called bystander effect, BE). We demonstrated that in the development of the BE an important role is played by oxidative stress (which is brought about by low radiation doses and ecDNA fragments of the culture medium of the irradiated cells), by an enzyme of apoptosis called caspase-3, and by DNA-binding receptors of the bystander cells, presumably TLR9. Proposed herein is a scheme of the development of an adaptive response and the BE on exposure to radiation. Ionizing radiation induces apoptosis of the radiosensitive fraction of cells due to the development of the "primary" oxidative stress (OS). DNA fragments of apoptotic cells are released into the intercellular space and interact with the DNA-binding receptors of the bystander cells. This interaction activates in lymphocytes signalling pathways associated with synthesis of the reactive oxygen species and nitrogen species, i.e., induces secondary oxidative stress accompanied by apoptosis of part of the cells, etc. Hence, single exposure to radiation may be followed by relatively long-lasting in the cellular population oxidative stress contributing to the development of an adaptive response. We thus believe that ecDNA of irradiated apoptotic lymphocytes is a significant factor of stress-signalling.
Collapse
Affiliation(s)
- Aleksei V Ermakov
- Research Centre for Medical Genetics, Russian Academy of Medical Science, Moscow, Russia.
| | | | | | | | | | | |
Collapse
|
15
|
Deng C, Shao Z, Xiong X, Liu Z, Zhang Z. The expression of TRAIL and its receptors in osteosarcoma cells and the apoptosis effect of a combination of TRAIL, adriamycin and IFN-γ on MG-63 cells. ACTA ACUST UNITED AC 2009. [DOI: 10.1016/s1007-4376(09)60065-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
16
|
Ogura A, Oowada S, Kon Y, Hirayama A, Yasui H, Meike S, Kobayashi S, Kuwabara M, Inanami O. Redox regulation in radiation-induced cytochrome c release from mitochondria of human lung carcinoma A549 cells. Cancer Lett 2009; 277:64-71. [DOI: 10.1016/j.canlet.2008.11.021] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Revised: 11/07/2008] [Accepted: 11/17/2008] [Indexed: 10/21/2022]
|
17
|
Abstract
The development of apoptosis resistance is a crucial step during the pathogenesis of malignant tumors. Thus, any treatment approach overcoming apoptosis resistance may be a valuable tool in oncology. Although a variety of treatments induce apoptosis, only very few specifically trigger programmed cell death. In this regard, the class of apoptosis inducing ligands may turn out to have a considerable potential in oncology. TNF-alpha-related apoptosis-inducing ligand (TRAIL/Apo2L) is the most promising candidate, either alone or in combination with established cancer therapies, since it induces apoptosis in a wide range of malignant cells while sparing most normal tissues. Since death-receptor induced apoptosis is mainly mediated via nonmitochondrial death pathways, it is possible to induce apoptosis in cancer cell systems which mainly harbor defects within the mitochondrial death cascades. Even more so it has been shown that conventional DNA damaging approaches reduced the killing threshold for receptor induced apoptosis, making TRAIL an ideal candidate for combined approaches. Thus, combined treatments might offer the chance to enhance therapeutic efficiency and overcome resistance. In combination, additive or synergistic apoptotic responses and substantially enhanced clonogenic cell kill has been documented. Furthermore, in several settings it has been shown that combined modality teatments were effective in malignant cells, which are highly resistant to either treatment, alone. Ionizing radiation is one of the most effective modalities in oncology. Thus, it is reasonable to test, how far combinations of TRAIL with ionizing radiation may increase the efficacy. Indeed, the combination of TRAIL with ionizing radiation in several in vitro settings as well as xenograft models resulted in highly increased rates of cell kill and long-term tumor control. No increase in the rate and severity of side effects has been documented, indicating that the combination really increases the therapeutic ratio. It is important to note that TRAIL and TRAIL receptor agonistic antibodies, either as single
Collapse
Affiliation(s)
- Olivier Niemöller
- Department of Radiation Oncology, Ludwig-Maximilians Universität Munich, Marchioninistr. 15, 81377 Munich, Germany
| | | |
Collapse
|
18
|
Pillai TG, Nair CKK, Janardhanan KK. Polysaccharides isolated from Ganoderma lucidum occurring in Southern parts of India, protects radiation induced damages both in vitro and in vivo. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2008; 26:80-85. [PMID: 21783892 DOI: 10.1016/j.etap.2008.02.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2007] [Revised: 01/31/2008] [Accepted: 02/05/2008] [Indexed: 05/31/2023]
Abstract
The in vivo and in vitro radioprotective property of the polysaccharides isolated from Ganoderma lucidum were determined by survival studies, induction of micronucleus in reticulocytes of mice, strand breaks in plasmid pBR322 DNA and inhibition of lipid peroxidation (TBARS assay). Polysaccharides were administered as a single dose after whole body exposure to 10Gy (60)Co γ-radiation to Swiss albino mice. At a dose of 500μg/kg body wt, the polysaccharides were most effective in protecting animals from radiation induced loss of lethality. Administration of 500μg/kg body wt to animal exposed to 10Gy gamma radiation resulted in more than 60% survival on the 30th day compared to the dose of 300mg/kg/body wt administration of amifostine, a clinically used radioprotective drug. The induction of micronuclei was reduced by the administration of polysaccharides. The decrease in micronuclei induction was dose dependent. Thus following 4Gy exposure the micronuclei in polychromatic erythrocytes (MNCE) was reduced from 28.16±3.049 to 16.0243±2.074 and 6.30±2.422 by polysaccharides at doses of 250μg/kg body wt and 500μg/kg body wt, respectively, and to 10.4±2.581 by amifostine at a dose of 300mg/kg body wt. The results indicate the significant protective effect of Ganoderma polysaccharides against radiation induced damages. The findings thus suggest the potential use of Ganoderma polysaccharides as novel radioprotective agent.
Collapse
Affiliation(s)
- Thulasi G Pillai
- Amala Cancer Research Centre, Amalanagar, Thrissur, Kerala, India
| | | | | |
Collapse
|
19
|
Takahashi M, Yasui H, Ogura A, Asanuma T, Kubota N, Tsujitani M, Kuwabara M, Inanami O. X irradiation combined with TNF alpha-related apoptosis-inducing ligand (TRAIL) reduces hypoxic regions of human gastric adenocarcinoma xenografts in SCID mice. JOURNAL OF RADIATION RESEARCH 2008; 49:153-161. [PMID: 18227621 DOI: 10.1269/jrr.07082] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Our previous study showed that X irradiation induced the expression of death receptor DR5 on the cell surface in tumor cell lines under not only normoxia but also hypoxia. X irradiation combined with TNF alpha-related apoptosis-inducing ligand (TRAIL), which is the ligand of DR5, induced apoptosis in vitro (Takahashi et al., (2007) Journal of Radiation Research, 48: 461-468). In this report, we examined the in vivo antitumor efficacy of X irradiation combined with TRAIL treatment in tumor xenograft models derived from human gastric adenocarcinoma MKN45 and MKN28 cells in SCID mice. X irradiation combined with TRAIL synergistically suppressed the tumor growth rates in the xenograft models derived from MKN45 and MKN28 cells, which have wild type Tp53 and mutated Tp53, respectively, indicating that the antitumor effects occurred in a Tp53-independent manner. Histological analysis showed that the combination of X irradiation and TRAIL induced caspase-3-dependent apoptotic cell death. Moreover, the immunohistochemical detection of hypoxic regions using the hypoxic marker pimonidazole revealed that caspase-3-dependent apoptosis occurred in the hypoxic regions in the tumors. These results indicated that X irradiation combined with TRAIL may be a useful treatment to reduce tumor growth in not only normoxic but also hypoxic regions.
Collapse
Affiliation(s)
- Momoko Takahashi
- Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Japan
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Sagan D, Mörtl S, Müller I, Eckardt-Schupp F, Eichholtz-Wirth H. Enhanced CD95-mediated apoptosis contributes to radiation hypersensitivity of NBS lymphoblasts. Apoptosis 2008; 12:753-67. [PMID: 17219051 DOI: 10.1007/s10495-006-0021-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The molecular causes for enhanced radiosensitivity of Nijmegen Breakage Syndrome cells are unclear, especially as repair of DNA damage is hardly impeded in these cells. We clearly demonstrate that radiation hypersensitivity is accompanied by enhanced gamma-radiation-induced apoptosis in NBS1 deficient lymphoblastoid cell lines. Differences in the apoptotic behavior of NBS1 (-/-) and NBS1 (+/-) cells are not due to an altered p53 stabilization or phosphorylation in NBS1 (-/-) cells. gamma-radiation-induced caspase-8 activity is increased and visualization of CD95 clustering by laser scanning microscopy shows a significant higher activation of the death receptor in NBS1 (-/-) cells. Further investigation of the molecular mechanisms reveals a role for reactive oxygen species-triggered activation of CD95. These results demonstrate that NBS1 suppresses the CD95 death receptor-dependent apoptotic pathway after gamma-irradiation and evidence is given that this is achieved by regulation of the PI3-K/AKT survival pathway.
Collapse
Affiliation(s)
- Daniel Sagan
- Institute of Radiobiology, GSF-National Research Center for Environment and Health, 85758 Neuherberg, Germany.
| | | | | | | | | |
Collapse
|
21
|
Takahashi M, Inanami O, Kubota N, Tsujitani M, Yasui H, Ogura A, Kuwabara M. Enhancement of cell death by TNF alpha-related apoptosis-inducing ligand (TRAIL) in human lung carcinoma A549 cells exposed to x rays under hypoxia. JOURNAL OF RADIATION RESEARCH 2007; 48:461-8. [PMID: 17895594 DOI: 10.1269/jrr.07028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Our previous study showed that ionizing radiation induced the expression of death receptor DR5 on the cell surface in tumor cell lines and that the death receptor of the TNF alpha-related apoptosis-inducing ligand TRAIL enhanced the apoptotic pathway (Hamasu et al., (2005) Journal of Radiation Research, 46:103-110). The present experiments were performed to examine whether treatment with TRAIL enhanced the cell killing in tumor cells exposed to ionizing radiation under hypoxia, since the presence of radioresistant cells in hypoxic regions of solid tumors is a serious problem in radiation therapy for tumors. When human lung carcinoma A549 cells were irradiated under normoxia and hypoxia, respectively, radiation-induced enhancement of expression of DR5 was observed under both conditions. Incubation in the presence of TRAIL enhanced the caspase-dependent and chymotrypsin-like-protease-dependent apoptotic cell death in A549 cells exposed to X rays. Furthermore, it was shown that treatment with TRAIL enhanced apoptotic cell death and loss of clonogenic ability in A549 cells exposed to X rays not only under normoxia but also under hypoxia, suggesting that combination treatment with TRAIL and X irradiation is effective for hypoxic tumor cells.
Collapse
Affiliation(s)
- Momoko Takahashi
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | |
Collapse
|
22
|
Marini P, Denzinger S, Schiller D, Kauder S, Welz S, Humphreys R, Daniel PT, Jendrossek V, Budach W, Belka C. Combined treatment of colorectal tumours with agonistic TRAIL receptor antibodies HGS-ETR1 and HGS-ETR2 and radiotherapy: enhanced effects in vitro and dose-dependent growth delay in vivo. Oncogene 2006; 25:5145-54. [PMID: 16636678 DOI: 10.1038/sj.onc.1209516] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We and others have demonstrated already that TRAIL (TNF-related apoptosis-inducing ligand) is a very promising candidate for molecular targeted anticancer therapy, especially when combined with ionizing radiation or other DNA-damaging agents. Agonist monoclonal antibodies that activate and are specific for the death signaling TRAIL receptors are an alternative method to stimulate the programmed cell death pathway. Phase 1 clinical trials have subsequently been conducted and shown a very good tolerability of these antibodies. In order to assess the efficacy of TRAIL receptor stimulation to induce cell death by this alternate method, we studied the combination of the agonistic-TRAIL receptor antibodies HGS-ETR1 and HGS-ETR2 with radiation in vitro and in vivo. Induction of apoptosis after combined treatment with TRAIL receptor antibodies HGS-ETR1 and/or HGS-ETR2 (0.01, 0.1, 1.0 mg/ml) and irradiation with 2, 5 or 10 Gy was determined by fluorescence microscopy and Western blot analysis of caspase-8 and PARP. The colorectal tumour cell lines Colo 205, HCT 116 and HCT-15 were used for in vitro experiments. Growth delay experiments were performed with combined treatment with fractionated irradiation (days 1-5 and 3 Gy single dose/day) and the receptor antibodies (intraperitonially, three different concentrations, application on days 1, 4 and 8) on Colo 205 xenograft-bearing NMRI (nu/nu) nude mice. HGS-ETR1 and HGS-ETR2 induced apoptotic cell death in a dose-dependent fashion and significantly increased cell death in combination with irradiation in vitro when compared to either irradiation or antibody treatment alone. The efficacy of the combined treatment seems to be at least partially Bax-dependent. Similar to the results from cell culture experiments, in vivo experiments demonstrated a dose-dependent delay in tumour growth after combined treatment. In vivo, in the Colo205 xenograft model, HGS-ETR2 revealed a higher activity than HGS-ETR1. This is the first study to demonstrate significant efficacy of combined treatment with the monoclonal agonistic TRAIL receptor antibodies HGS-ETR1 and HGS-ETR2 and ionising radiation in in vitro and in vivo models. We postulate that HGS-ETR1 and HGS-ETR2 will be very promising new agents in the field of molecular targeted multi-modality anticancer therapy.
Collapse
Affiliation(s)
- P Marini
- CCC Tübingen, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|