201
|
Casimiro S, Alho I, Bettencourt M, Pires R, Lipton A, Costa L. RANKL enhances the effect of an antagonist of inhibitor of apoptosis proteins (cIAPs) in RANK-positive breast cancer cells. J Bone Oncol 2013; 2:116-22. [PMID: 26909281 PMCID: PMC4723389 DOI: 10.1016/j.jbo.2013.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 06/05/2013] [Accepted: 07/01/2013] [Indexed: 11/16/2022] Open
Abstract
Objective Between 65% and 75% of patients with metastatic breast cancer will have decreased 5-year survival and increased morbidity due to cancer relapse in bone. At this stage of disease treatment is palliative, but tumor-targeted compounds could add to the benefits of anti-resorptive agents, improving clinical outcome. Inhibitor-of-apoptosis proteins (IAPs) are overexpressed in many tumors and second mitochondria-derived activator of caspases (Smac) mimetics have been designed to antagonize IAPs. In this work we explored the use of AT-406, a Smac mimetic, to target the tumor compartment of bone metastases. Methods Effect of AT-406 on cancer cells apoptosis, expression of IAPs and osteogenic potential was addressed in vitro using the RANK-positive MDA-MB-231 breast cancer cell line. Effect of AT-406 on osteoclastogenesis was determined by inducing the differentiation of the RAW 264.7 mouse monocytic cell line. Osteoclastogenesis was measured by TRAP staining and TRACP 5b quantification. Results AT-406 increased apoptosis in MDA-MB-231 breast cancer cells in vitro, and activation of RANK-pathway improved this effect. RANKL stimuli induced a strong increase in c-IAP2. AT-406 increased osteoclast differentiation and activity, by up-regulating the osteogenic transcription factor Nfatc1, but also increased the apoptosis of mature osteoclasts in the absence of RANKL. Conclusions Our results indicate that despite the anti-tumoral effect of AT-406, its use in the context of bone metastatic disease needs to be carefully monitored for the induction of increased bone resorption. We also hypothesize that the combination of AT-406 with anti-RANKL directed therapies could have a beneficial effect, especially in RANK-positive tumors.
Collapse
Affiliation(s)
- S Casimiro
- Clinical and Translational Oncology Research Unit, Institute of Molecular Medicine, Lisbon Medical School, Lisbon, Portugal
| | - I Alho
- Clinical and Translational Oncology Research Unit, Institute of Molecular Medicine, Lisbon Medical School, Lisbon, Portugal
| | - M Bettencourt
- Clinical and Translational Oncology Research Unit, Institute of Molecular Medicine, Lisbon Medical School, Lisbon, Portugal
| | - R Pires
- Clinical and Translational Oncology Research Unit, Institute of Molecular Medicine, Lisbon Medical School, Lisbon, Portugal
| | - A Lipton
- Penn State Hershey Medical Center, Hershey, PA, USA
| | - L Costa
- Clinical and Translational Oncology Research Unit, Institute of Molecular Medicine, Lisbon Medical School, Lisbon, Portugal; Oncology Department, Hospital de Santa Maria-CHLN, Lisbon, Portugal
| |
Collapse
|
202
|
Farrand L, Byun S, Kim JY, Im-Aram A, Lee J, Lim S, Lee KW, Suh JY, Lee HJ, Tsang BK. Piceatannol enhances cisplatin sensitivity in ovarian cancer via modulation of p53, X-linked inhibitor of apoptosis protein (XIAP), and mitochondrial fission. J Biol Chem 2013; 288:23740-50. [PMID: 23833193 DOI: 10.1074/jbc.m113.487686] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Resistance to cisplatin (CDDP) in ovarian cancer (OVCA) arises from the dysregulation of tumor suppressors and survival signals. During genotoxic challenge, these factors can be influenced by secondary agents that facilitate the induction of apoptosis. Piceatannol is a natural metabolite of the stilbene resveratrol found in grapes and is converted from its parent compound by the enzyme CYP1BA1 p450. It has been hypothesized to exert specific effects against various cellular targets; however, its ability to influence CDDP resistance in cancer cells has not been investigated to date. Here, we show that piceatannol is a potent enhancer of CDDP sensitivity in OVCA, and this effect is achieved through the modulation of several major determinants of chemoresistance. Piceatannol enhances p53-mediated expression of the pro-apoptotic protein NOXA, increases XIAP degradation via the ubiquitin-proteasome pathway, and enhances caspase-3 activation. This response is associated with an increase in Drp1-dependent mitochondrial fission, leading to more effective induction of apoptosis. In vivo studies using a mouse model of OVCA reveal that a number of these changes occur in association with a greater overall reduction in tumor weight when mice are treated with both piceatannol and CDDP, in comparison to treatment with either agent alone. Taken together, these findings demonstrate the potential application of piceatannol to enhance CDDP sensitivity in OVCA, and it acts on p53, XIAP, and mitochondrial fission.
Collapse
Affiliation(s)
- Lee Farrand
- World Class University Major in Biomodulation, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Parrish AB, Freel CD, Kornbluth S. Cellular mechanisms controlling caspase activation and function. Cold Spring Harb Perspect Biol 2013; 5:5/6/a008672. [PMID: 23732469 DOI: 10.1101/cshperspect.a008672] [Citation(s) in RCA: 445] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Caspases are the primary drivers of apoptotic cell death, cleaving cellular proteins that are critical for dismantling the dying cell. Initially translated as inactive zymogenic precursors, caspases are activated in response to a variety of cell death stimuli. In addition to factors required for their direct activation (e.g., dimerizing adaptor proteins in the case of initiator caspases that lie at the apex of apoptotic signaling cascades), caspases are regulated by a variety of cellular factors in a myriad of physiological and pathological settings. For example, caspases may be modified posttranslationally (e.g., by phosphorylation or ubiquitylation) or through interaction of modulatory factors with either the zymogenic or active form of a caspase, altering its activation and/or activity. These regulatory events may inhibit or enhance enzymatic activity or may affect activity toward particular cellular substrates. Finally, there is emerging literature to suggest that caspases can participate in a variety of cellular processes unrelated to apoptotic cell death. In these settings, it is particularly important that caspases are maintained under stringent control to avoid inadvertent cell death. It is likely that continued examination of these processes will reveal new mechanisms of caspase regulation with implications well beyond control of apoptotic cell death.
Collapse
Affiliation(s)
- Amanda B Parrish
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
204
|
Activation of executioner caspases is a predictor of progression-free survival in glioblastoma patients: a systems medicine approach. Cell Death Dis 2013; 4:e629. [PMID: 23681224 PMCID: PMC3674364 DOI: 10.1038/cddis.2013.157] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. GBM cells are highly resistant to apoptosis induced by antitumor drugs and radiotherapy resulting in cancer progression. We assessed whether a systems medicine approach, analysing the ability of tumor cells to execute apoptosis could be utilized to predict the response of GBM patients to treatment. Concentrations of the key proapoptotic proteins procaspase-3, procaspase-9, Smac and Apaf-1 and the antiapopotic protein XIAP were determined in a panel of GBM cell lines and GBM patient tumor resections. These values were used as input for APOPTO-CELL, a systems biological based mathematical model built to predict cellular susceptibility to undergo caspase activation. The modeling was capable of accurately distinguishing between GBM cells that die or survive in response to treatment with temozolomide in 10 of the 11 lines analysed. Importantly the results obtained using GBM patient samples show that APOPTO-CELL was capable of stratifying patients according to their progression-free survival times and predicted the ability of tumor cells to support caspase activation in 16 of the 21 GBM patients analysed. Calculating the susceptibility to apoptosis execution may be a potent tool in predicting GBM patient therapy responsiveness and may allow for the use of APOPTO-CELL in a clinical setting.
Collapse
|
205
|
Luo LF, Hou CC, Yang WX. Nuclear factors: roles related to mitochondrial deafness. Gene 2013; 520:79-89. [PMID: 23510774 DOI: 10.1016/j.gene.2013.03.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 03/08/2013] [Indexed: 12/16/2022]
Abstract
Hearing loss (HL) is a common disorder with mitochondrial dysfunction as one of the major causes leading to deafness. Mitochondrial dysfunction may be caused by either mutations in nuclear genes leading to defective nuclear-encoded proteins or mutations in mitochondrial genes leading to defective mitochondrial-encoded products. The specific nuclear genes involved in HL can be classified into two categories depending on whether mitochondrial gene mutations co-exist (modifier genes) or not (deafness-causing genes). TFB1M, MTO1, GTPBP3, and TRMU are modifier genes. A mutation in any of these modifier genes may lead to a deafness phenotype when accompanied by the mitochondrial gene mutation. OPA1, TIMM8A, SMAC/DIABLO, MPV17, PDSS1, BCS1L, SUCLA2, C10ORF2, COX10, PLOG1and RRM2B are deafness-causing genes. A mutation in any of these deafness-causing genes will directly induce variable phenotypic HL.
Collapse
Affiliation(s)
- Ling-Feng Luo
- Institute of Cell and Developmental Biology, Zhejiang University, Hangzhou 310058, China
| | | | | |
Collapse
|
206
|
Abstract
The number of available eukaryotic genomes has expanded to the point where we can evaluate the complete evolutionary history of many cellular processes. Such analyses for the apoptosis regulatory networks suggest that this network already existed in the ancestor of the entire animal kingdom (Metazoa) in a form more complex than in some popular animal model organisms. This supports the growing realization that regulatory networks do not necessarily evolve from simple to complex and that the relative simplicity of these networks in nematodes and insects does not represent an ancestral state, but is the result of secondary simplifications. Network evolution is not a process of monotonous increase in complexity, but a dynamic process that includes lineage-specific gene losses and expansions, protein domain reshuffling, and emergence/reemergence of similar protein architectures by parallel evolution. Studying the evolution of such networks is a challenging yet interesting subject for research and investigation, and such studies on the apoptosis networks provide us with interesting hints of how these networks, critical in so many human diseases, have developed.
Collapse
Affiliation(s)
- Christian M Zmasek
- Program on Bioinformatics and Systems Biology, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | | |
Collapse
|
207
|
Ng KB, Bustamam A, Sukari MA, Abdelwahab SI, Mohan S, Buckle MJC, Kamalidehghan B, Nadzri NM, Anasamy T, A Hadi AH, Rahman HS. Induction of selective cytotoxicity and apoptosis in human T4-lymphoblastoid cell line (CEMss) by boesenbergin a isolated from boesenbergia rotunda rhizomes involves mitochondrial pathway, activation of caspase 3 and G2/M phase cell cycle arrest. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 13:41. [PMID: 23432947 PMCID: PMC3600682 DOI: 10.1186/1472-6882-13-41] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Accepted: 02/14/2013] [Indexed: 11/18/2022]
Abstract
Background Boesenbergia rotunda (Roxb.) Schlecht (family zingiberaceae) is a rhizomatous herb that is distributed from north-eastern India to south-east Asia, especially in Indonesia, Thailand and Malaysia. Previous research has shown that the crude extract of this plant has cytotoxic properties. The current study examines the cytotoxic properties of boesenbergin A isolated from Boesenbergia rotunda. Methods MTT assay was used to check the cytotoxicity of boesenbergin A. The morphological assessment of apoptosis was monitored using normal and fluorescence microscopy. The early and late phase of apoptosis was investigated using annexin V and DNA laddering assays, respectively. The mitochondrial membrane potential (MMP) was assessed by fluorescence microscopy. Human apoptosis proteome profiler assays were performed to investigate the mechanism of cell death. In addition, the protein levels of Bax, Bcl2 and HSP 70 were also analyzed using western blot. Assays of caspase =-3/7, -8 and =-9 were carried out in order to test for induction during treatment. Lastly, cell cycle progression was analyzed using flow cytometry. Results Boesenbergin A was found to have the highest toxicity towards CEMss cancer cells (IC50 = 8 μg/ml). The morphology of CEMss cells after treatment showed evidence of apoptosis that included blebbing and chromatin condensation. The annexin V assay revealed that early apoptosis is induced after treatment. The DNA laddering assay confirmed that DNA fragmentation had occurred during late apoptosis. The cell cycle analysis indicated that boesenbergin A was able to induce G2/M phase arrest in CEMss cells. The activity of caspases -3/7, -8 and -9 was increased after treatment which indicates both intrinsic and extrinsic pathways are induced during apoptosis. The involvement of mitochondria was established by increased mitochondrial membrane potential and up and down regulation of Bcl2 and Bax proteins as well as HSP70. Conclusion In conclusion, the results demonstrated that boesenbergin A induced apoptosis of CEMss cells through Bcl2/Bax signaling pathways with the involvement of caspases and G2/M phase cell cycle arrest. The current findings warrant further research on boesenbergin A as a novel chemotherapeutic agent for leukemia intervention including studies in animal models.
Collapse
|
208
|
Govender R, Phulukdaree A, Gengan RM, Anand K, Chuturgoon AA. Silver nanoparticles of Albizia adianthifolia: the induction of apoptosis in human lung carcinoma cell line. J Nanobiotechnology 2013; 11:5. [PMID: 23418790 PMCID: PMC3606379 DOI: 10.1186/1477-3155-11-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 01/30/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Silver nanoparticles (AgNP), the most popular nano-compounds, possess unique properties. Albizia adianthifolia (AA) is a plant of the Fabaceae family that is rich in saponins. The biological properties of a novel AgNP, synthesized from an aqueous leaf extract of AA (AA(AgNP)), were investigated on A549 lung cells. Cell viability was determined by the MTT assay. Cellular oxidative status (lipid peroxidation and glutathione (GSH) levels), ATP concentration, caspase-3/-7, -8 and -9 activities were determined. Apoptosis, mitochondrial (mt) membrane depolarization (flow cytometry) and DNA fragmentation (comet assay) were assessed. The expression of CD95 receptors, p53, bax, PARP-1 and smac/DIABLO was evaluated by flow cytometry and/or western blotting. RESULTS Silver nanoparticles of AA caused a dose-dependent decrease in cell viability with a significant increase in lipid peroxidation (5-fold vs. control; p = 0.0098) and decreased intracellular GSH (p = 0.1184). A significant 2.5-fold decrease in cellular ATP was observed upon AA(AgNP) exposure (p = 0.0040) with a highly significant elevation in mt depolarization (3.3-fold vs. control; p < 0.0001). Apoptosis was also significantly higher (1.5-fold) in AA(AgNP) treated cells (p < 0.0001) with a significant decline in expression of CD95 receptors (p = 0.0416). Silver nanoparticles of AA caused a significant 2.5-fold reduction in caspase-8 activity (p = 0.0024) with contrasting increases in caspase-3/-7 (1.7-fold vs. control; p = 0.0180) and -9 activity (1.4-fold vs. control; p = 0.0117). Western blots showed increased expression of smac/DIABLO (4.1-fold) in treated cells (p = 0.0033). Furthermore, AA(AgNP) significantly increased the expression of p53, bax and PARP-1 (1.2-fold; p = 0.0498, 1.6-fold; p = 0.0083 and 1.1-fold; p = 0.0359 respectively). CONCLUSION Data suggests that AA(AgNP) induces cell death in the A549 lung cells via the mt mediated intrinsic apoptotic program. Further investigation is required to potentiate the use of this novel compound in cancer therapy trials.
Collapse
Affiliation(s)
- Rishalan Govender
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Private Bag 7, Congella, Durban, 4013, South Africa
| | | | | | | | | |
Collapse
|
209
|
Govender R, Phulukdaree A, Gengan RM, Anand K, Chuturgoon AA. Silver nanoparticles of Albizia adianthifolia: the induction of apoptosis in human lung carcinoma cell line. J Nanobiotechnology 2013. [PMID: 23418790 DOI: 10.1186/1477–3155–11–5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Silver nanoparticles (AgNP), the most popular nano-compounds, possess unique properties. Albizia adianthifolia (AA) is a plant of the Fabaceae family that is rich in saponins. The biological properties of a novel AgNP, synthesized from an aqueous leaf extract of AA (AA(AgNP)), were investigated on A549 lung cells. Cell viability was determined by the MTT assay. Cellular oxidative status (lipid peroxidation and glutathione (GSH) levels), ATP concentration, caspase-3/-7, -8 and -9 activities were determined. Apoptosis, mitochondrial (mt) membrane depolarization (flow cytometry) and DNA fragmentation (comet assay) were assessed. The expression of CD95 receptors, p53, bax, PARP-1 and smac/DIABLO was evaluated by flow cytometry and/or western blotting. RESULTS Silver nanoparticles of AA caused a dose-dependent decrease in cell viability with a significant increase in lipid peroxidation (5-fold vs. control; p = 0.0098) and decreased intracellular GSH (p = 0.1184). A significant 2.5-fold decrease in cellular ATP was observed upon AA(AgNP) exposure (p = 0.0040) with a highly significant elevation in mt depolarization (3.3-fold vs. control; p < 0.0001). Apoptosis was also significantly higher (1.5-fold) in AA(AgNP) treated cells (p < 0.0001) with a significant decline in expression of CD95 receptors (p = 0.0416). Silver nanoparticles of AA caused a significant 2.5-fold reduction in caspase-8 activity (p = 0.0024) with contrasting increases in caspase-3/-7 (1.7-fold vs. control; p = 0.0180) and -9 activity (1.4-fold vs. control; p = 0.0117). Western blots showed increased expression of smac/DIABLO (4.1-fold) in treated cells (p = 0.0033). Furthermore, AA(AgNP) significantly increased the expression of p53, bax and PARP-1 (1.2-fold; p = 0.0498, 1.6-fold; p = 0.0083 and 1.1-fold; p = 0.0359 respectively). CONCLUSION Data suggests that AA(AgNP) induces cell death in the A549 lung cells via the mt mediated intrinsic apoptotic program. Further investigation is required to potentiate the use of this novel compound in cancer therapy trials.
Collapse
Affiliation(s)
- Rishalan Govender
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Private Bag 7, Congella, Durban, 4013, South Africa
| | | | | | | | | |
Collapse
|
210
|
Chen F, Xu C, Du L, Wang Y, Cao J, Fu Y, Guo Y, Liu Q, Fan F. Tat-SmacN7 induces radiosensitization in cancer cells through the activation of caspases and induction of apoptosis. Int J Oncol 2013; 42:985-92. [PMID: 23338568 DOI: 10.3892/ijo.2013.1785] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 12/21/2012] [Indexed: 11/05/2022] Open
Abstract
A major concern in cancer therapy is resistance of tumors such as human non-small cell lung cancer and esophageal cancer to radiotherapy. Intrinsic radioresistance of these cancer cells limits therapeutic efficiency. Here, we determined in two cancer cell lines the potential radiosensitizing activity of Tat-SmacN7, a small molecule compound, which mimics the activity of Smac, a mitochondrial protein released during apoptosis. We found that Tat-SmacN7 can enter the cells and promote RNA expression and the activity of caspase-3, -8 and -9 and sensitized the cancer cells to radiation with a sensitization enhancement ratio (SER) of 1.5-1.6. Tat-SmacN7 radiosensitization was mediated by both extrinsic and intrinsic apoptosis pathways through activation of caspases. Consistently, blockage of caspase activation, through treatment with a caspase inhibitor, z-VAD-fmk, inhibited apoptosis and abrogated Tat-SmacN7 radiosensitization. Our study demonstrates that Tat-SmacN7 also has radiosensitization effects in vivo, so it could be further developed as a novel class of radiosensitizers for the treatment of radioresistant human non-small cell lung cancer and esophageal cancer.
Collapse
Affiliation(s)
- Fenghua Chen
- Department of Radiation Hazard Evaluation, Institute of Radiation Medicine of Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
211
|
Abstract
The description of apoptosis and the identification of the genes that regulate it have proved pivotal to our understanding of how cancer cells accumulate and ultimately cause morbidity and mortality. It has become increasingly clear that in CLL the balance between the pro- and anti-apoptotic members of the BCL2 family of apoptotic regulatory proteins is critical in the development and clinical progression of CLL. Furthermore, the apoptotic potential of the CLL cell determines chemotherapy sensitivity and ultimately progression-free and overall survival. The unravelling of the BCL2 story in CLL has led to the development of a whole new class of therapeutic agents-the BH3 mimetics-which are significantly more targeted than conventional chemo-immunotherapy and therefore promise potent clinical activity coupled with reduced toxicity.
Collapse
|
212
|
Wang S, Bai L, Lu J, Liu L, Yang CY, Sun H. Targeting inhibitors of apoptosis proteins (IAPs) for new breast cancer therapeutics. J Mammary Gland Biol Neoplasia 2012; 17:217-28. [PMID: 23054134 PMCID: PMC3534930 DOI: 10.1007/s10911-012-9265-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 08/07/2012] [Indexed: 01/07/2023] Open
Abstract
Apoptosis resistance is a hallmark of human cancer. Research in the last two decades has identified key regulators of apoptosis, including inhibitor of apoptosis proteins (IAPs). These critical apoptosis regulators have been targeted for the development of new cancer therapeutics. In this article, we will discuss three members of IAP proteins, namely XIAP, cIAP1 and cIAP2, as cancer therapeutic targets and the progress made in developing new cancer therapeutic agents to target these IAP proteins.
Collapse
Affiliation(s)
- Shaomeng Wang
- Comprehensive Cancer Center and Departments of Internal Medicine, Pharmacology and Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | | | | | |
Collapse
|
213
|
MAO HONGLUAN, PANG YINGXIN, ZHANG XIAOLEI, YANG FANG, ZHENG JINGFANG, WANG YU, LIU PEISHU. Smac peptide potentiates TRAIL- or paclitaxel-mediated ovarian cancer cell death in vitro and in vivo. Oncol Rep 2012; 29:515-22. [DOI: 10.3892/or.2012.2132] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 09/28/2012] [Indexed: 11/06/2022] Open
|
214
|
Zhou B, Zhang J, Chen G, You L, Zhang TP, Zhao YP. Therapy of Smac mimetic SM-164 in combination with gemcitabine for pancreatic cancer. Cancer Lett 2012; 329:118-24. [PMID: 23142291 DOI: 10.1016/j.canlet.2012.10.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 10/22/2012] [Accepted: 10/27/2012] [Indexed: 11/27/2022]
Abstract
Pancreatic cancer (PC) is a lethal solid malignancy with resistance to traditional chemotherapy. We investigated therapy of PC with SM-164 and gemcitabine alone and in combination. Survival of PC cells was reduced as the dose of SM-164 increased. SM-164 and/or gemcitabine increased the number of apoptotic and dead PC cells, and expression of cleavage fragments of caspase-3 and PARP1, and inhibited tumor xenograft growth in nude mice. The inhibitory effect of combination treatment was greater and of longer duration than monotherapy. Neither combination nor monotherapy showed any significant toxicity in vivo. Apoptosis and necrosis, decreased expression of Ki67, and increased expression of cleaved caspase-3 were observed in xenograft tumor tissues in SM164/gemcitabine-treated mice. SM-164 could be a promising new agent for treatment of PC in combination with gemcitabine.
Collapse
Affiliation(s)
- Bin Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | | | | |
Collapse
|
215
|
Manzoni L, Belvisi L, Bianchi A, Conti A, Drago C, de Matteo M, Ferrante L, Mastrangelo E, Perego P, Potenza D, Scolastico C, Servida F, Timpano G, Vasile F, Rizzo V, Seneci P. Homo- and heterodimeric Smac mimetics/IAP inhibitors as in vivo-active pro-apoptotic agents. Part I: Synthesis. Bioorg Med Chem 2012; 20:6687-708. [DOI: 10.1016/j.bmc.2012.09.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Revised: 09/05/2012] [Accepted: 09/11/2012] [Indexed: 12/22/2022]
|
216
|
Hegde M, Karki SS, Thomas E, Kumar S, Panjamurthy K, Ranganatha SR, Rangappa KS, Choudhary B, Raghavan SC. Novel levamisole derivative induces extrinsic pathway of apoptosis in cancer cells and inhibits tumor progression in mice. PLoS One 2012; 7:e43632. [PMID: 22970136 PMCID: PMC3438185 DOI: 10.1371/journal.pone.0043632] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 07/23/2012] [Indexed: 11/23/2022] Open
Abstract
Background Levamisole, an imidazo(2,1-b)thiazole derivative, has been reported to be a potential antitumor agent. In the present study, we have investigated the mechanism of action of one of the recently identified analogues, 4a (2-benzyl-6-(4′-fluorophenyl)-5-thiocyanato-imidazo[2,1-b][1], [3], [4]thiadiazole). Materials and Methods ROS production and expression of various apoptotic proteins were measured following 4a treatment in leukemia cell lines. Tumor animal models were used to evaluate the effect of 4a in comparison with Levamisole on progression of breast adenocarcinoma and survival. Immunohistochemistry and western blotting studies were performed to understand the mechanism of 4a action both ex vivo and in vivo. Results We have determined the IC50 value of 4a in many leukemic and breast cancer cell lines and found CEM cells most sensitive (IC50 5 µM). Results showed that 4a treatment leads to the accumulation of ROS. Western blot analysis showed upregulation of pro-apoptotic proteins t-BID and BAX, upon treatment with 4a. Besides, dose-dependent activation of p53 along with FAS, FAS-L, and cleavage of CASPASE-8 suggest that it induces death receptor mediated apoptotic pathway in CEM cells. More importantly, we observed a reduction in tumor growth and significant increase in survival upon oral administration of 4a (20 mg/kg, six doses) in mice. In comparison, 4a was found to be more potent than its parental analogue Levamisole based on both ex vivo and in vivo studies. Further, immunohistochemistry and western blotting studies indicate that 4a treatment led to abrogation of tumor cell proliferation and activation of apoptosis by the extrinsic pathway even in animal models. Conclusion Thus, our results suggest that 4a could be used as a potent chemotherapeutic agent.
Collapse
Affiliation(s)
- Mahesh Hegde
- Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Subhas S. Karki
- Department of Pharmaceutical Chemistry, KLE University's College of Pharmacy, Bangalore, Karnataka, India
| | - Elizabeth Thomas
- Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Sujeet Kumar
- Department of Pharmaceutical Chemistry, KLE University's College of Pharmacy, Bangalore, Karnataka, India
| | - Kuppusamy Panjamurthy
- Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | | | | | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bangalore, Karnataka, India
| | - Sathees C. Raghavan
- Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka, India
- * E-mail:
| |
Collapse
|
217
|
Application of flow cytometry to determine differential redistribution of cytochrome c and Smac/DIABLO from mitochondria during cell death signaling. PLoS One 2012; 7:e42298. [PMID: 22848756 PMCID: PMC3407092 DOI: 10.1371/journal.pone.0042298] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 07/05/2012] [Indexed: 12/02/2022] Open
Abstract
Mitochondrially mediated apoptosis is characterized by redistribution of proteins from mitochondria to cytoplasm following permeabilization of the outer mitochondrial membrane. We applied flow cytometry to quantify simultaneously the redistribution of two apoptogenic proteins, cytochrome c (cyt c) and Smac/DIABLO (Smac). Mammalian cells were treated with digitonin that selectively permeabilizes the plasma membrane. Following fixation, treated cells were infused successively with primary and secondary antibodies (the latter fluorescently tagged) enabling independent detection of cyt c and Smac. Digitonin-treated cells that retain cyt c or Smac in mitochondria generate strong fluorescence signals in flow cytometry. Cells in which cyt c or Smac have transited the outer mitochondrial membrane show greatly reduced fluorescence because the proteins are lost from the digitonin-permeabilized cells. Quantitative flow cytometry revealed that in 143B TK- cells treated with staurosporine, cyt c and Smac exit mitochondria asymmetrically, with cyt c redistribution preceding that of Smac. However, in HeLa cells likewise treated, cyt c and Smac exit mitochondria concurrently. Under other conditions of apoptotic induction, for example, 143B TK- cells treated with MT-21 (an apoptotic inducer that binds to the mitochondrial adenine nucleotide transporter), redistribution of Smac precedes that of cyt c. The various patterns of redistribution of these proteins were confirmed by immunocytochemical analysis and confocal microscopy. We conclude that flow cytometry can be employed effectively to quantify simultaneously the redistribution of cyt c and Smac from mitochondria to the cytosol. Moreover, differential redistribution of cyt c and Smac occurs under various conditions, thereby reflecting constraints on availability of these proteins to exit mitochondria after permeabilization of the outer membrane.
Collapse
|
218
|
Wang SB, Tan Y, Lei W, Wang YG, Zhou XM, Jia XY, Zhang KJ, Chu L, Liu XY, Qian WB. Complete eradication of xenograft hepatoma by oncolytic adenovirus ZD55 harboring TRAIL-IETD-Smac gene with broad antitumor effect. Hum Gene Ther 2012; 23:992-1002. [PMID: 22530834 DOI: 10.1089/hum.2011.159] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Cancer-targeting dual-gene virotherapy (CTGVT-DG) is an important modification of CTGVT, in which two suitable genes are used to obtain an excellent antitumor effect. A key problem is to join the two genes to form one fused gene, and then to clone it into the oncolytic viral vector so that only one investigational new drug application, instead of two, is required for clinical use. Many linkers (e.g., internal ribosome entry site) are used to join two genes together, but they are not all equally efficacious. Here, we describe finding the best linker, that is, sequence encoding the four amino acids IETD, to join the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene and the second mitochondria-derived activator of caspase (Smac) gene to form TRAIL-IETD-Smac and inserting it into oncolytic viral vector ZD55 to construct ZD55-TRAIL-IETD-Smac, which matched ZD55-TRAIL plus ZD55-Smac in completely eliminating xenograft hepatoma. ZD55-TRAIL-IETD-Smac works by quantitative cleavage at IETD↓by inducing caspase-8; activation or inhibition of caspase-8 could up- or downregulate cleavage, respectively. The cleaved product, TRAIL-IETD, does not affect the function of TRAIL. Numerous experiments have shown that the combined use of ZD55-TRAIL plus ZD55-X could completely eradicate many xenograft tumors, and therefore the IETD is potentially a useful linker to construct many antitumor drugs, for example, ZD55-TRAIL-IETD-X, where X has a compensative or synergetic effect on TRAIL. We found that the antitumor effect of ZD55-IL-24-IETD-TRAIL also has an equivalent antitumor effect compared with the combined use of ZD55-IL-24 plus ZD55-TRAIL, because ZD55-IL-24 could also induce caspase-8. This means that IETD, as a two-gene linker, may have broad use.
Collapse
Affiliation(s)
- Shi-Bing Wang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
219
|
Tang P, Huang H. Progress in understanding the role of inhibitor of apoptosis proteins in molecular targeted therapy of esophageal cancer. Shijie Huaren Xiaohua Zazhi 2012; 20:1843-1847. [DOI: 10.11569/wcjd.v20.i20.1843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The apoptosis and antiapoptotic signaling pathways play a critical role in the embryonic and lymphocyte development, immune system modulation, and tissue homeostasis, as well as carcinogenesis. As inhibitor of apoptosis proteins (IAPs) are highly expressed in several neoplasms and are closely related to carcinogenesis, cancer progression, radiochemotherapeutic resistance, and prognosis, therapies targeting IAPs have become a research hotspot for molecular targeted therapy of tumors. In recent years, many agents targeting IAPs which are being evaluated in clinical trials are showing promising prospect for neoplastic therapy. As such, the identification of key roles of IAPs in esophageal cancer has revealed their potential value as therapeutic targets. This report reviews the progress in understanding the role of IAPs in molecular targeted therapy of esophageal cancer.
Collapse
|
220
|
Quast SA, Berger A, Buttstädt N, Friebel K, Schönherr R, Eberle J. General Sensitization of melanoma cells for TRAIL-induced apoptosis by the potassium channel inhibitor TRAM-34 depends on release of SMAC. PLoS One 2012; 7:e39290. [PMID: 22723988 PMCID: PMC3377761 DOI: 10.1371/journal.pone.0039290] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 05/20/2012] [Indexed: 11/18/2022] Open
Abstract
The death ligand TRAIL represents a promising therapeutic strategy for metastatic melanoma, however prevalent and inducible resistance limit its applicability. A new approach is presented here for sensitization to TRAIL. It is based on inhibition of the membrane potassium channel KCa3.1 (IK1), which serves fundamental cellular functions related to membrane potential. The selective inhibitor TRAM-34 did not induce apoptosis by itself but synergistically enhanced TRAIL sensitivity and overrode TRAIL resistance in a large panel of melanoma cell lines. Expression of IK1 was also found in mitochondria, and its inhibition resulted in mitochondrial membrane hyperpolarization and an early activation of Bax. The combination of TRAM-34 and TRAIL resulted in massive release of mitochondrial factors, cytochrome c, AIF and SMAC/DIABLO. Bax knockdown and Bcl-2 overexpression abolished apoptosis. Overexpression of XIAP diminished apoptosis by two-fold, and SMAC knockdown almost completely abolished apoptosis. These data uncover the existence of a rheostat in melanoma cells, consisting of inhibitor of apoptosis proteins and SMAC, which regulates TRAIL sensitivity. Thus, a new strategy is described based on mitochondrial membrane channels, which correspond to Bax activation. As both TRAIL and IK1 inhibitors had shown only minor side effects in clinical trials, a clinical application of this combination is conceivable.
Collapse
Affiliation(s)
- Sandra-Annika Quast
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
- Institute for Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
| | - Anja Berger
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| | - Nicole Buttstädt
- Department of Biophysics, Center of Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Kristin Friebel
- Department of Biophysics, Center of Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Roland Schönherr
- Department of Biophysics, Center of Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Jürgen Eberle
- Department of Dermatology and Allergy, Skin Cancer Center, University Medical Center Charité, Berlin, Germany
| |
Collapse
|
221
|
Targeting the Apo2L/TRAIL system for the therapy of autoimmune diseases and cancer. Biochem Pharmacol 2012; 83:1475-83. [DOI: 10.1016/j.bcp.2011.12.036] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 12/22/2011] [Accepted: 12/22/2011] [Indexed: 01/07/2023]
|
222
|
Abstract
Evading programmed cell death is one of the hallmarks of cancer. Conversely, inducing cell death by pharmacological means is the basis of almost every non-invasive cancer therapy. Research over the past decade has greatly increased our understanding of non-apoptotic programmed cell death events, such as lysosomal-mediated cell death, necroptosis and cell death with autophagy. It is becoming clear that an intricate effector network connects many of these classical and non-classical death pathways. In this Review, we discuss converging and diverging features of these pathways, as well as attempts to exploit this newly gained knowledge pharmacologically to provide therapeutics for cancer.
Collapse
Affiliation(s)
- Peter Kreuzaler
- University of Cambridge, Department of Pathology, Tennis Court Road, Cambridge CB2 1QP, UK
| | | |
Collapse
|
223
|
Chen KF, Lin JP, Shiau CW, Tai WT, Liu CY, Yu HC, Chen PJ, Cheng AL. Inhibition of Bcl-2 improves effect of LCL161, a SMAC mimetic, in hepatocellular carcinoma cells. Biochem Pharmacol 2012; 84:268-77. [PMID: 22580047 DOI: 10.1016/j.bcp.2012.04.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/18/2012] [Accepted: 04/30/2012] [Indexed: 02/02/2023]
Abstract
In this study, we investigated the effect of LCL161, a SMAC mimetic, in hepatocellular carcinoma (HCC). LCL161 showed differential effects on apoptosis in four HCC cell lines, and the endogenous level of Bcl-2 determined the sensitivity of HCC cells to LCL161. Cytotoxicity and apoptosis were observed in sensitive PLC5 and Hep3B cells that express lower levels of Bcl-2, but not in resistant Huh-7 and SK-Hep1 cells with higher Bcl-2 expression. Down regulation of Bcl-2 by small interference RNA overcame the resistance to LCL161 in Huh-7, and the apoptotic effect was rescued in Bcl-2-expressing Hep3B. To test the hypothesis that Bcl-2 determines the sensitivity of HCC cells to LCL161, we assayed the biological effect of SC-2001, a novel Bcl-2 inhibitor derived from obatoclax, in LCL161-resistant cell lines. Huh-7 cells co-treated with LCL161 and SC-2001 showed a significant dose-dependent apoptotic effect demonstrated by sub-G1 assay and cleavage of PARP. Furthermore, the combination index (CI) of LCL161 and SC-2001 showed a convincing synergism in resistant Huh-7. In addition, the combinational therapy showed significant growth inhibition in Huh-7-bearing xenograft tumors. Notably, down regulation of Bcl-2 was observed in a tumor sample treated with LCL161 and SC-2001. In conclusion, targeting Bcl-2 with SC-2001 overcomes drug resistance to LCL161 in HCC cells thus suggesting a new anti-IAP combinational therapy for HCC.
Collapse
Affiliation(s)
- Kuen-Feng Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
224
|
Perier C, Bové J, Vila M. Mitochondria and programmed cell death in Parkinson's disease: apoptosis and beyond. Antioxid Redox Signal 2012; 16:883-95. [PMID: 21619488 DOI: 10.1089/ars.2011.4074] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
UNLABELLED Abstract Significance: Activation of mitochondrion-dependent programmed cell death (PCD) pathways is instrumental to the demise of substantia nigra pars compacta dopaminergic neurons in experimental mouse models of Parkinson's disease (PD). Supporting the relevance of these findings for PD, key molecular elements of this pathogenic cascade have also been demonstrated in postmortem brain samples of PD patients. Recent Advances and Critical Issues: Mounting evidence indicates that different morphological types of cell death co-exist in the brain of PD patients, all of which may result from the activation of common upstream PCD pathways. Indeed, contrary to initial views, it is now established that the deleterious effects of PCD pathways are not limited to mitochondrion-mediated caspase-dependent apoptosis but also involve caspase-independent nonapoptotic cell death, including necrosis. This notion may help reconcile the observation of both apoptotic and nonapoptotic dopaminergic cell death in postmortem PD samples. FUTURE DIRECTIONS Potential neuroprotective strategies for PD should be aimed at targeting both apoptotic and nonapoptotic pathways, all of which may simultaneously occur in PD patients through activation of common upstream PCD pathways involving the mitochondria. Antioxid. Redox Signal. 16, 883-895.
Collapse
Affiliation(s)
- Celine Perier
- Vall d'Hebron Research Institute-CIBERNED, Barcelona, Spain
| | | | | |
Collapse
|
225
|
Zacchino V, Minghetti M, Centoducati G, Leaver MJ. Diablo/SMAC: a novel biomarker of pollutant exposure in European flounder (Platichthys flesus). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2012; 79:176-183. [PMID: 22244825 DOI: 10.1016/j.ecoenv.2011.12.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 12/20/2011] [Accepted: 12/28/2011] [Indexed: 05/31/2023]
Abstract
Diablo (or SMAC) is a protein released from mitochondria following apoptotic stimuli and inhibits the actions of Inhibitors of Apoptosis (IAP) proteins. IAPs regulate the activity of caspases and NFkB, the primary executioners of apoptosis and of inflammation, respectively. Thus, Diablo is important for the regulation of cellular responses to damage. In Northern Europe, statutory governmental marine monitoring programs measure various biomarkers in flounder to indicate biological effects of pollutant exposure. More recently transcriptomic techniques have been applied in flounder to gain a more comprehensive understanding of pollutant effects, and to discover novel biomarkers. In most of these studies utilising flounder, Diablo was amongst the most highly increased transcripts identified. The aim of this study was to further examine piscine Diablo, at the gene level and mRNA level, after exposure to prototypical pollutants, and in flounder caught from polluted environments. The results show that two genes encoding Diablo exist in fish species, and in flounder one of these genes is increased in liver after exposure to polyaromatic hydrocarbons and polychlorinated biphenyls, and also in livers from fish living on contaminated estuarine sediments. Therefore, Diablo measurement has potential as a biomarker of pollutant exposure, and could indicate damaging effects of chemical contaminants.
Collapse
Affiliation(s)
- Valentina Zacchino
- Institute of Aquaculture, University of Stirling, Stirling FK9 4LA, UK; Dipartimento di Sanità Pubblica e Zootecnia, Università di Bari, Italy
| | - Matteo Minghetti
- Institute of Aquaculture, University of Stirling, Stirling FK9 4LA, UK
| | | | - Michael J Leaver
- Institute of Aquaculture, University of Stirling, Stirling FK9 4LA, UK.
| |
Collapse
|
226
|
Peptides for cancer therapy: a drug-development opportunity and a drug-delivery challenge. Ther Deliv 2012; 3:609-21. [DOI: 10.4155/tde.12.37] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Therapeutic peptides (TPs) are a class of peptide-based agents capable of eliciting a therapeutic response by modulation of targets within or on the surface of cells. TPs are advantageous because they are amenable to rational design, they have high specificity for their targets and can be made to target almost any protein of interest, including proteins for which we have no small-molecule drugs. Owing to this versatility, TPs have a great potential for cancer therapy in an age of personalized medicine, in which we need novel drugs to target the many novel pathways being discovered as tumor drivers. However, in order to utilize TPs as drugs, many obstacles must be overcome. TPs have short half-lives in systemic circulation, are easily degraded by proteases in plasma and target cells, are often cleared by the reticuloendothelial system and can be immunogenic. This article will discuss ways of overcoming many of these hurdles by utilizing macromolecular peptide delivery systems and tumor-targeting agents.
Collapse
|
227
|
Houghton PJ, Kang MH, Reynolds CP, Morton CL, Kolb EA, Gorlick R, Keir ST, Carol H, Lock R, Maris JM, Billups CA, Smith MA. Initial testing (stage 1) of LCL161, a SMAC mimetic, by the Pediatric Preclinical Testing Program. Pediatr Blood Cancer 2012; 58:636-9. [PMID: 21681929 PMCID: PMC3253328 DOI: 10.1002/pbc.23167] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 03/28/2011] [Indexed: 11/05/2022]
Abstract
LCL161, a SMAC mimetic, was tested against the PPTP in vitro panel (1.0 nM to 10.0 µM) and the PPTP in vivo panels (30 or 75 mg/kg [solid tumors] or 100 mg/kg [ALL]) administered orally twice in a week. LCL161 showed a median relative IC(50) value of >10 µM, being more potent against several leukemia and lymphoma lines. In vivo LCL161 induced significant differences in EFS distribution in approximately one-third of solid tumor xenografts (osteosarcoma and glioblastoma), but not in ALL xenografts. No objective tumor responses were observed. In vivo LCL161 demonstrated limited single agent activity against the pediatric preclinical models studied.
Collapse
|
228
|
Sun H, Tawa G, Wallqvist A. Classification of scaffold-hopping approaches. Drug Discov Today 2012; 17:310-24. [PMID: 22056715 PMCID: PMC3328312 DOI: 10.1016/j.drudis.2011.10.024] [Citation(s) in RCA: 271] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 10/17/2011] [Accepted: 10/19/2011] [Indexed: 10/15/2022]
Abstract
The general goal of drug discovery is to identify novel compounds that are active against a preselected biological target with acceptable pharmacological properties defined by marketed drugs. Scaffold hopping has been widely applied by medicinal chemists to discover equipotent compounds with novel backbones that have improved properties. In this article we classify scaffold hopping into four major categories, namely heterocycle replacements, ring opening or closure, peptidomimetics and topology-based hopping. We review the structural diversity of original and final scaffolds with respect to each category. We discuss the advantages and limitations of small, medium and large-step scaffold hopping. Finally, we summarize software that is frequently used to facilitate different kinds of scaffold-hopping methods.
Collapse
Affiliation(s)
- Hongmao Sun
- Biotechnology HPC Software Applications Institute, Telemedicine and Advanced Technology Research Center, US Army Medical Research and Materiel Command, Fort Frederick, MD 21702, USA.
| | | | | |
Collapse
|
229
|
Flygare JA, Beresini M, Budha N, Chan H, Chan IT, Cheeti S, Cohen F, Deshayes K, Doerner K, Eckhardt SG, Elliott LO, Feng B, Franklin MC, Reisner SF, Gazzard L, Halladay J, Hymowitz SG, La H, LoRusso P, Maurer B, Murray L, Plise E, Quan C, Stephan JP, Young SG, Tom J, Tsui V, Um J, Varfolomeev E, Vucic D, Wagner AJ, Wallweber HJA, Wang L, Ware J, Wen Z, Wong H, Wong JM, Wong M, Wong S, Yu R, Zobel K, Fairbrother WJ. Discovery of a potent small-molecule antagonist of inhibitor of apoptosis (IAP) proteins and clinical candidate for the treatment of cancer (GDC-0152). J Med Chem 2012; 55:4101-13. [PMID: 22413863 DOI: 10.1021/jm300060k] [Citation(s) in RCA: 187] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A series of compounds were designed and synthesized as antagonists of cIAP1/2, ML-IAP, and XIAP based on the N-terminus, AVPI, of mature Smac. Compound 1 (GDC-0152) has the best profile of these compounds; it binds to the XIAP BIR3 domain, the BIR domain of ML-IAP, and the BIR3 domains of cIAP1 and cIAP2 with K(i) values of 28, 14, 17, and 43 nM, respectively. These compounds promote degradation of cIAP1, induce activation of caspase-3/7, and lead to decreased viability of breast cancer cells without affecting normal mammary epithelial cells. Compound 1 inhibits tumor growth when dosed orally in the MDA-MB-231 breast cancer xenograft model. Compound 1 was advanced to human clinical trials, and it exhibited linear pharmacokinetics over the dose range (0.049 to 1.48 mg/kg) tested. Mean plasma clearance in humans was 9 ± 3 mL/min/kg, and the volume of distribution was 0.6 ± 0.2 L/kg.
Collapse
Affiliation(s)
- John A Flygare
- Department of Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Domingues C, Ryoo HD. Drosophila BRUCE inhibits apoptosis through non-lysine ubiquitination of the IAP-antagonist REAPER. Cell Death Differ 2012; 19:470-7. [PMID: 21886178 PMCID: PMC3235231 DOI: 10.1038/cdd.2011.116] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 07/29/2011] [Accepted: 07/29/2011] [Indexed: 11/10/2022] Open
Abstract
Active caspases execute apoptosis to eliminate superfluous or harmful cells in animals. In Drosophila, living cells prevent uncontrolled caspase activation through an inhibitor of apoptosis protein (IAP) family member, dIAP1, and apoptosis is preceded by the expression of IAP-antagonists, such as Reaper, Hid and Grim. Strong genetic modifiers of this pathway include another IAP family gene encoding an E2 ubiquitin conjugating enzyme domain, dBruce. Although the genetic effects of dBruce mutants are well documented, molecular targets of its encoded protein have remained elusive. Here, we report that dBruce targets Reaper for ubiquitination through an unconventional mechanism. Specifically, we show that dBruce physically interacts with Reaper, dependent upon Reaper's IAP-binding (IBM) and GH3 motifs. Consistently, Reaper levels were elevated in a dBruce -/- background. Unexpectedly, we found that dBruce also affects the levels of a mutant form of Reaper without any internal lysine residues, which normally serve as conventional ubiquitin acceptor sites. Furthermore, we were able to biochemically detect ubiquitin conjugation on lysine-deficient Reaper proteins, and knockdown of dBruce significantly reduced the extent of this ubiquitination. Our results indicate that dBruce inhibits apoptosis by promoting IAP-antagonist ubiquitination on unconventional acceptor sites.
Collapse
Affiliation(s)
- C Domingues
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - H D Ryoo
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
231
|
Feltham R, Khan N, Silke J. IAPS and ubiquitylation. IUBMB Life 2012; 64:411-8. [DOI: 10.1002/iub.565] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 07/25/2011] [Indexed: 11/11/2022]
|
232
|
Inhibitor of Apoptosis (IAP) proteins as therapeutic targets for radiosensitization of human cancers. Cancer Treat Rev 2012; 38:760-6. [PMID: 22342104 DOI: 10.1016/j.ctrv.2012.01.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Revised: 01/17/2012] [Accepted: 01/23/2012] [Indexed: 01/22/2023]
Abstract
Radiotherapy initiates a variety of signaling events in cancer cells that eventually lead to cell death in case the DNA damage cannot be repaired. However, the signal transduction pathways that mediate cell death in response to radiation-inflicted DNA damage are frequently disturbed in human cancers, contributing to radioresistance. For example, aberrant activation of antiapoptotic programs such as high expression of Inhibitor of Apoptosis (IAP) proteins has been shown to interfere with the efficacy of radiotherapy. Since IAP proteins have been linked to radioresistance in several malignancies, therapeutic targeting of IAP proteins may open new perspectives to overcome radioresistance. Therefore, molecular targeting of IAP proteins may provide novel opportunities to reactivate cell death pathways that mediate radiation-induced cytotoxicity. A number of strategies have been developed in recent years to antagonize IAP proteins for the treatment of cancers. Some of these approaches have already been translated into a clinical application. While IAP protein-targeting agents are currently being evaluated in early clinical trials alone or in combination with conventional chemotherapy, they have not yet been tested in combination with radiation therapy. Therefore, it is a timely subject to discuss the opportunities of antagonizing IAP proteins for radiosensitization. Preclinical studies demonstrating the potential of this concept in relevant in vitro and in vivo models underscore that this combination approach warrants further clinical investigation. Thus, combination protocols using IAP antagonists together with radiotherapy may pave the avenue to more effective radiation-based treatment options for cancer patients.
Collapse
|
233
|
Bianchi A, Ugazzi M, Ferrante L, Lecis D, Scavullo C, Mastrangelo E, Seneci P. Rational design, synthesis and characterization of potent, drug-like monomeric Smac mimetics as pro-apoptotic anticancer agents. Bioorg Med Chem Lett 2012; 22:2204-8. [PMID: 22342627 DOI: 10.1016/j.bmcl.2012.01.098] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 01/24/2012] [Accepted: 01/25/2012] [Indexed: 11/15/2022]
Abstract
A set of phenyl-substituted Smac mimetics/IAP inhibitor analogues of lead compound 2a was synthesized, aiming to retain its strong cell-free potency while increasing its bioavailability. Seventeen compounds 2b-r were prepared and characterized in vitro, using cell-free and cellular assays. Among them, the p-CF(3) substituted analogue 2m showed the best permeability through cell membranes, and was selected for further in vitro and in vivo studies due to its strong, sub-micromolar cellular potency.
Collapse
Affiliation(s)
- Aldo Bianchi
- CISI scrl, Via Fantoli 16/15, I-20138 Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
234
|
Fulda S, Vucic D. Targeting IAP proteins for therapeutic intervention in cancer. Nat Rev Drug Discov 2012; 11:109-24. [PMID: 22293567 DOI: 10.1038/nrd3627] [Citation(s) in RCA: 641] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Evasion of apoptosis is one of the crucial acquired capabilities used by cancer cells to fend off anticancer therapies. Inhibitor of apoptosis (IAP) proteins exert a range of biological activities that promote cancer cell survival and proliferation. X chromosome-linked IAP is a direct inhibitor of caspases - pro-apoptotic executioner proteases - whereas cellular IAP proteins block the assembly of pro-apoptotic protein signalling complexes and mediate the expression of anti-apoptotic molecules. Furthermore, mutations, amplifications and chromosomal translocations of IAP genes are associated with various malignancies. Among the therapeutic strategies that have been designed to target IAP proteins, the most widely used approach is based on mimicking the IAP-binding motif of second mitochondria-derived activator of caspase (SMAC), which functions as an endogenous IAP antagonist. Alternative strategies include transcriptional repression and the use of antisense oligonucleotides. This Review provides an update on IAP protein biology as well as current and future perspectives on targeting IAP proteins for therapeutic intervention in human malignancies.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe University Frankfurt, Komturstr. 3a, 60528 Frankfurt, Germany.
| | | |
Collapse
|
235
|
Fulda S. Exploiting inhibitor of apoptosis proteins as therapeutic targets in hematological malignancies. Leukemia 2012; 26:1155-65. [PMID: 22230799 DOI: 10.1038/leu.2012.4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Resistance to apoptosis is one of the hallmarks of human cancers and contributes to the insensitivity of many cancers to commonly used treatment approaches. Inhibitor of apoptosis (IAP) proteins, a family of anti-apoptotic proteins, have an important role in evasion of apoptosis, as they can both block apoptosis-signaling pathways and promote survival. High expression of IAP proteins is observed in multiple cancers, including hematological malignancies, and has been associated with unfavorable prognosis and poor patients' outcome. Therefore, IAP proteins are currently considered as promising molecular targets for therapy. Indeed, drug-discovery approaches over the last decade aiming at neutralizing IAP proteins have resulted in the generation of small-molecule inhibitors or antisense oligonucleotides that demonstrated in vitro and in vivo antitumor activities in preclinical studies. As some of these strategies have already entered the stage of clinical evaluation, for example, in leukemia, an update on this promising molecular-targeted strategy to interfere with apoptotic pathways is of broad interest.
Collapse
Affiliation(s)
- S Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany.
| |
Collapse
|
236
|
Smac: Its role in apoptosis induction and use in lung cancer diagnosis and treatment. Cancer Lett 2012; 318:9-13. [PMID: 22227574 DOI: 10.1016/j.canlet.2011.12.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Revised: 12/11/2011] [Accepted: 12/13/2011] [Indexed: 01/23/2023]
Abstract
Apoptosis is a conserved and regulated cell suicide process, the malfunction of which is closely linked with carcinogenesis. Caspases control the induction of apoptosis through an enzymatic cascade that can be activated by both the mitochondrial and death receptor pathways. Smac is a mitochondrial protein that interacts with Inhibitor of Apoptosis Proteins (IAPs) and, upon apoptotic stimuli, is released into the cytoplasm to inhibit the capase-binding activity of IAPs. Smac plays key roles in both the diagnosis and treatment of cancer, especially lung cancer. Our review will focus on the roles of Smac in lung carcinogenesis and cancer progression and its relevance in lung cancer treatment.
Collapse
|
237
|
Potenza D, Belvisi L, Vasile F, Moroni E, Cossu F, Seneci P. A NMR and computational study of Smac mimics targeting both the BIR2 and BIR3 domains in XIAP protein. Org Biomol Chem 2012; 10:3278-87. [DOI: 10.1039/c2ob06979b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
238
|
Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, Dawson TM, Dawson VL, El-Deiry WS, Fulda S, Gottlieb E, Green DR, Hengartner MO, Kepp O, Knight RA, Kumar S, Lipton SA, Lu X, Madeo F, Malorni W, Mehlen P, Nuñez G, Peter ME, Piacentini M, Rubinsztein DC, Shi Y, Simon HU, Vandenabeele P, White E, Yuan J, Zhivotovsky B, Melino G, Kroemer G. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ 2012; 19:107-20. [PMID: 21760595 PMCID: PMC3252826 DOI: 10.1038/cdd.2011.96] [Citation(s) in RCA: 1871] [Impact Index Per Article: 143.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 06/13/2011] [Indexed: 02/07/2023] Open
Abstract
In 2009, the Nomenclature Committee on Cell Death (NCCD) proposed a set of recommendations for the definition of distinct cell death morphologies and for the appropriate use of cell death-related terminology, including 'apoptosis', 'necrosis' and 'mitotic catastrophe'. In view of the substantial progress in the biochemical and genetic exploration of cell death, time has come to switch from morphological to molecular definitions of cell death modalities. Here we propose a functional classification of cell death subroutines that applies to both in vitro and in vivo settings and includes extrinsic apoptosis, caspase-dependent or -independent intrinsic apoptosis, regulated necrosis, autophagic cell death and mitotic catastrophe. Moreover, we discuss the utility of expressions indicating additional cell death modalities. On the basis of the new, revised NCCD classification, cell death subroutines are defined by a series of precise, measurable biochemical features.
Collapse
Affiliation(s)
- L Galluzzi
- INSERM U848, ‘Apoptosis, Cancer and Immunity', 94805 Villejuif, France
- Institut Gustave Roussy, 94805 Villejuif, France
- Université Paris Sud-XI, 94805 Villejuif, France
| | - I Vitale
- INSERM U848, ‘Apoptosis, Cancer and Immunity', 94805 Villejuif, France
- Institut Gustave Roussy, 94805 Villejuif, France
- Université Paris Sud-XI, 94805 Villejuif, France
| | - J M Abrams
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - E S Alnemri
- Department of Biochemistry and Molecular Biology, Center for Apoptosis Research, Kimmel Cancer Institute, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - E H Baehrecke
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - M V Blagosklonny
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - T M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - V L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - W S El-Deiry
- Cancer Institute Penn State, Hershey Medical Center, Philadelphia, PA 17033, USA
| | - S Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe University, Frankfurt 60528, Germany
| | - E Gottlieb
- The Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - D R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - M O Hengartner
- Institute of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland
| | - O Kepp
- INSERM U848, ‘Apoptosis, Cancer and Immunity', 94805 Villejuif, France
- Institut Gustave Roussy, 94805 Villejuif, France
- Université Paris Sud-XI, 94805 Villejuif, France
| | - R A Knight
- Institute of Child Health, University College London, London WC1N 3JH, UK
| | - S Kumar
- Centre for Cancer Biology, SA Pathology, Adelaide, South Australia 5000, Australia
- Department of Medicine, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - S A Lipton
- Sanford-Burnham Medical Research Institute, San Diego, CA 92037, USA
- Salk Institute for Biological Studies, , La Jolla, CA 92037, USA
- The Scripps Research Institute, La Jolla, CA 92037, USA
- Univerisity of California, San Diego, La Jolla, CA 92093, USA
| | - X Lu
- Ludwig Institute for Cancer Research, Oxford OX3 7DQ, UK
| | - F Madeo
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - W Malorni
- Department of Therapeutic Research and Medicines Evaluation, Section of Cell Aging and Degeneration, Istituto Superiore di Sanità, 00161 Rome, Italy
- Istituto San Raffaele Sulmona, 67039 Sulmona, Italy
| | - P Mehlen
- Apoptosis, Cancer and Development, CRCL, 69008 Lyon, France
- INSERM, U1052, 69008 Lyon, France
- CNRS, UMR5286, 69008 Lyon, France
- Centre Léon Bérard, 69008 Lyon, France
| | - G Nuñez
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - M E Peter
- Northwestern University Feinberg School of Medicine, Chicago, IL 60637, USA
| | - M Piacentini
- Laboratory of Cell Biology, National Institute for Infectious Diseases IRCCS ‘L Spallanzani', 00149 Rome, Italy
- Department of Biology, University of Rome ‘Tor Vergata', 00133 Rome, Italy
| | - D C Rubinsztein
- Cambridge Institute for Medical Research, Cambridge CB2 0XY, UK
| | - Y Shi
- Shanghai Institutes for Biological Sciences, 200031 Shanghai, China
| | - H-U Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - P Vandenabeele
- Department for Molecular Biology, Gent University, 9052 Gent, Belgium
- Department for Molecular Biomedical Research, VIB, 9052 Gent, Belgium
| | - E White
- The Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - J Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - B Zhivotovsky
- Institute of Environmental Medicine, Division of Toxicology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - G Melino
- Biochemical Laboratory IDI-IRCCS, Department of Experimental Medicine, University of Rome ‘Tor Vergata', 00133 Rome, Italy
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
| | - G Kroemer
- INSERM U848, ‘Apoptosis, Cancer and Immunity', 94805 Villejuif, France
- Metabolomics Platform, Institut Gustave Roussy, 94805 Villejuif, France
- Centre de Recherche des Cordeliers, 75005 Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75908 Paris, France
- Université Paris Descartes, Paris 5, 75270 Paris, France
| |
Collapse
|
239
|
MIYOSHI T, NAKANO SI, NAKAMURA K, YAMANOUCHI K, NISHIHARA M. In Vivo Electroporation Induces Cell Cycle Reentry of Myonuclei in Rat Skeletal Muscle. J Vet Med Sci 2012; 74:1291-7. [DOI: 10.1292/jvms.12-0195] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Takahiro MIYOSHI
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1–1–1 Yayoi, Bunkyo-ku, Tokyo 113–8657, Japan
| | - Shin-ichi NAKANO
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1–1–1 Yayoi, Bunkyo-ku, Tokyo 113–8657, Japan
| | - Katsuyuki NAKAMURA
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1–1–1 Yayoi, Bunkyo-ku, Tokyo 113–8657, Japan
| | - Keitaro YAMANOUCHI
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1–1–1 Yayoi, Bunkyo-ku, Tokyo 113–8657, Japan
| | - Masugi NISHIHARA
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1–1–1 Yayoi, Bunkyo-ku, Tokyo 113–8657, Japan
| |
Collapse
|
240
|
|
241
|
Xu Y, Xin Y, Diao Y, Lu C, Fu J, Luo L, Yin Z. Synergistic effects of apigenin and paclitaxel on apoptosis of cancer cells. PLoS One 2011; 6:e29169. [PMID: 22216199 PMCID: PMC3244456 DOI: 10.1371/journal.pone.0029169] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 11/22/2011] [Indexed: 01/20/2023] Open
Abstract
Background It was well known that the clinical use of chemotherapeutic drugs is restricted by severe adverse reactions and drug resistances. Thus it is necessary to figure out a strategy to increase the specific anti-tumor efficiency of chemotherapeutic drugs. Apigenin, a kind of flavonoids, has been reported to possess anticancer activities with very low cytotoxicity to normal tissue. Methodology/Principal Findings Our results from cell viability assay, western-blots and TdT-mediated dUTP-biotin nick end labeling (TUNEL) assay demonstrated the synergistic pro-apoptotic effects of a low dose of apigenin and paclitaxel in human cancer cell lines. To analyze the underlying mechanism, we examined reactive oxygen species (ROS) staining after cells were treated with a combination of apigenin and paclitaxel, or each of them alone. Data from flow-cytometry showed that superoxides but not reduction of peroxides accumulated in HeLa cells treated with apigenin or a combination of apigenin and paclitaxel. Apigenin and paclitaxel-induced HeLa cell apoptosis was related to the level of ROS in cells. We further evaluated activity and protein level of superoxide dismutase (SOD). Apigenin significantly inhibited SOD activity but did not alter the SOD protein level suggesting that apigenin promoted ROS accumulation through suppressing enzyme activity of SOD. Addition of Zn2+, Cu2+ and Mn2+ to cell lysates inhibited apigenin's effects on SOD activity. At the same time, data from caspase-2 over-expression and knocked-down experiments demonstrated that caspase-2 participated in apigenin and paclitaxel-induced HeLa cell apoptosis. Conclusions/Significance Taken together, our study demonstrated that apigenin can sensitize cancer cells to paclitaxel induced apoptosis through suppressing SOD activity, which then led to accumulation of ROS and cleavage of caspase-2, suggesting that the combined use of apigenin and paclitaxel was an effective way to decrease the dose of paclitaxel taken.
Collapse
Affiliation(s)
- Yimiao Xu
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Yinqiang Xin
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Ying Diao
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Changyan Lu
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Jin Fu
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, People's Republic of China
- * E-mail: (ZY); (LL)
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
- * E-mail: (ZY); (LL)
| |
Collapse
|
242
|
Phillipps HR, Kokay IC, Grattan DR, Hurst PR. X-linked inhibitor of apoptosis protein and active caspase-3 expression patterns in antral follicles in the sheep ovary. Reproduction 2011; 142:855-67. [DOI: 10.1530/rep-11-0177] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
X-linked inhibitor of apoptosis protein (XIAP) interacts with caspases to inhibit their activity, thereby providing a potential mechanism for regulation of granulosa cell apoptosis occurring during follicular atresia. The aim of this study was to determine the presence and localization of XIAP mRNA and protein content in the sheep ovary and compare these expression patterns with active caspase-3 protein in the same antral follicles. Romney ewe estrous cycles (n=25) were synchronized with 2–3 Estrumate injections and ovarian tissue collected during the luteal and follicular phases of the cycle. The presence ofXIAPmRNA was confirmed by RT-PCR using laser capture microdissected ovarian cell samples.XIAPmRNA was subsequently localized byin situhybridization histochemistry and XIAP and active caspase-3 protein visualized by immunohistochemistry. In antral follicles extensive XIAP localization was evident in both granulosa and thecal cells. In contrast, mRNA expression was widespread in granulosa cells and only detected in thecal tissue from a small proportion of antral follicles. Active caspase-3 and XIAP comparative expression analysis showed positiveXIAPmRNA expression in all late luteal phase (day 14) follicles, despite varying levels of active caspase-3 protein. A proportion of follicular phase (days 15 and 16) follicles, however, showed an inverse expression relationship at the protein and mRNA levels in both granulosa and thecal tissue, as did XIAP protein in day 14 follicles. These results suggest high XIAP may prevent activation of caspase-3, thereby regulating follicular atresia in antral follicles and could potentially be utilized as a marker of follicular health.
Collapse
|
243
|
LING BAOPING, ZHANG RUI, WANG ZHIGUO, LIU YONGJUN, LIU CHENGBU. STUDY ON THE INTERACTIONS OF Smac MIMETICS WITH XIAP-BIR3 DOMAIN BY DOCKING AND MOLECULAR DYNAMICS SIMULATIONS. JOURNAL OF THEORETICAL & COMPUTATIONAL CHEMISTRY 2011. [DOI: 10.1142/s0219633610005980] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Upon receiving an apoptotic stimulus, the mature mitochondrial protein second mitochondria-derived activator of caspases (Smac)/direct IAP-binding protein with low PI (DIABLO), which could be released from mitochondria into the cytosol together with cytochrome C , specifically binds to inhibitor of apoptosis proteins (IAPs) and relieves the inhibitory effect of caspase, thus promotes cell death. Some artificial small molecules (called Smac mimetics) can mimic the N-terminal four residues Ala1-Val2-Pro3-Ile4 (AVPI) sequence of mitochondrial protein Smac, and competitively bind to X-linked inhibitor of apoptosis protein baculoviral IAP repeats (XIAP-BIR3) domain with caspase-9, which leads to the removal of the inhibition of caspase-9 by XIAP and induce apoptosis. To gain an insight into the nature of XIAP-BIR3 domain recognizing Smac mimetics, we used docking and molecular dynamics simulations methods to study four representative Smac mimetics. The docking results show that the orientations of these backbones of ligands are identical with that of AVPI in the binding pocket. Each ligand corresponds to two competitive conformations, which are called extended and bended conformations. The results of molecular dynamics simulations show that the extended conformation is more stable, and the calculations of energy decomposition reveal that the residue Thr308 makes the strongest interaction with XIAP-BIR3. In addition, Asp309, Glu314, and Trp323 are indispensable for XIAP-BIR3 recognizing and binding Smac mimetics.
Collapse
Affiliation(s)
- BAOPING LING
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, P. R. China
| | - RUI ZHANG
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810001, P. R. China
| | - ZHIGUO WANG
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810001, P. R. China
| | - YONGJUN LIU
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, P. R. China
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810001, P. R. China
| | - CHENGBU LIU
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, P. R. China
| |
Collapse
|
244
|
Le Quement ST, Ishoey M, Petersen MT, Thastrup J, Hagel G, Nielsen TE. Solid-phase synthesis of smac peptidomimetics incorporating triazoloprolines and biarylalanines. ACS COMBINATORIAL SCIENCE 2011; 13:667-75. [PMID: 21905744 DOI: 10.1021/co200078u] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Apoptotic induction mechanisms are of crucial importance for the general homeostasis of multicellular organisms. In cancer the apoptotic pathways are downregulated, which, at least partly, is due to an abundance of inhibitors of apoptosis proteins (IAPs) that block the apoptotic cascade by deactivating proteolytic caspases. The Smac protein has an antagonistic effect on IAPs, thus providing structural clues for the synthesis of new pro-apoptotic compounds. Herein, we report a solid-phase approach for the synthesis of Smac-derived tetrapeptide libraries. On the basis of a common (N-Me)AVPF sequence, peptides incorporating triazoloprolines and biarylalanines were synthesized by means of Cu(I)-catalyzed azide-alkyne cycloaddition and Pd-catalyzed Suzuki cross-coupling reactions. Solid-phase procedures were optimized to high efficiency, thus accessing all products in excellent crude purities and yields (both typically above 90%). The peptides were subjected to biological evaluation in a live/dead cellular assay which revealed that structural decorations on the AVPF sequence indeed are highly important for cytotoxicity toward HeLa cells.
Collapse
Affiliation(s)
| | - Mette Ishoey
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Mette T. Petersen
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Jacob Thastrup
- 2CureX, Department of Surgery, K, Bispebjerg Hospital, Bispebjerg Bakke 23, DK-2400 Copenhagen NV, Denmark
| | - Grith Hagel
- 2CureX, Department of Surgery, K, Bispebjerg Hospital, Bispebjerg Bakke 23, DK-2400 Copenhagen NV, Denmark
| | - Thomas E. Nielsen
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| |
Collapse
|
245
|
The role of curcumin on intestinal oxidative stress, cell proliferation and apoptosis after ischemia/reperfusion injury in rats. J Mol Histol 2011; 42:579-87. [DOI: 10.1007/s10735-011-9364-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Accepted: 09/26/2011] [Indexed: 12/12/2022]
|
246
|
Reingewertz TH, Shalev DE, Sukenik S, Blatt O, Rotem-Bamberger S, Lebendiker M, Larisch S, Friedler A. Mechanism of the interaction between the intrinsically disordered C-terminus of the pro-apoptotic ARTS protein and the Bir3 domain of XIAP. PLoS One 2011; 6:e24655. [PMID: 21949740 PMCID: PMC3176765 DOI: 10.1371/journal.pone.0024655] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 08/15/2011] [Indexed: 11/18/2022] Open
Abstract
ARTS (Sept4_i2) is a mitochondrial pro-apoptotic protein that functions as a tumor suppressor. Its expression is significantly reduced in leukemia and lymphoma patients. ARTS binds and inhibits XIAP (X-linked Inhibitor of Apoptosis protein) by interacting with its Bir3 domain. ARTS promotes degradation of XIAP through the proteasome pathway. By doing so, ARTS removes XIAP inhibition of caspases and enables apoptosis to proceed. ARTS contains 27 unique residues in its C-terminal domain (CTD, residues 248–274) which are important for XIAP binding. Here we characterized the molecular details of this interaction. Biophysical and computational methods were used to show that the ARTS CTD is intrinsically disordered under physiological conditions. Direct binding of ARTS CTD to Bir3 was demonstrated using NMR and fluorescence spectroscopy. The Bir3 interacting region in ARTS CTD was mapped to ARTS residues 266–274, which are the nine C-terminal residues in the protein. Alanine scan of ARTS 266–274 showed the importance of several residues for Bir3 binding, with His268 and Cys273 contributing the most. Adding a reducing agent prevented binding to Bir3. A dimer of ARTS 266–274 formed by oxidation of the Cys residues into a disulfide bond bound with similar affinity and was probably required for the interaction with Bir3. The detailed analysis of the ARTS – Bir3 interaction provides the basis for setting it as a target for anti cancer drug design: It will enable the development of compounds that mimic ARTS CTD, remove IAPs inhibition of caspases, and thereby induce apoptosis.
Collapse
Affiliation(s)
- Tali H. Reingewertz
- Institute of Chemistry, Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem, Israel
| | - Deborah E. Shalev
- The Wolfson Centre for Applied Structural Biology, Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem, Israel
| | - Shahar Sukenik
- Institute of Chemistry, Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem, Israel
| | - Ofrah Blatt
- Institute of Chemistry, Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem, Israel
| | - Shahar Rotem-Bamberger
- Institute of Chemistry, Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem, Israel
| | - Mario Lebendiker
- The Wolfson Centre for Applied Structural Biology, Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem, Israel
| | - Sarit Larisch
- Cell Death Research Laboratory, Department of Biology, Faculty of Natural Sciences, University of Haifa, Mount Carmel, Haifa, Israel
| | - Assaf Friedler
- Institute of Chemistry, Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
247
|
Smac-mimetic compound SM-164 induces radiosensitization in breast cancer cells through activation of caspases and induction of apoptosis. Breast Cancer Res Treat 2011; 133:189-99. [PMID: 21901386 DOI: 10.1007/s10549-011-1752-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 08/18/2011] [Indexed: 01/15/2023]
Abstract
Radiotherapy is a treatment choice for local control of breast cancer, particularly after the removal of tumor tissues by surgery. However, intrinsic radioresistance of cancer cells limits therapeutic efficacy. Here, we determined in breast cancer cells the potential radiosensitizing activity of SM-164, a small molecule compound, that mimics the activity of SMAC, a mitochondrial protein released during apoptosis to activate caspases by inhibiting cellular inhibitor of apoptosis proteins, cIAP-1, and XIAP. We found that SM-164 at nanomolar concentrations promoted degradation of cIAP-1, disrupted the inhibitory binding of XIAP to active caspase-9, and sensitized breast cancer cells to radiation with a sensitization enhancement ratio (SER) of 1.7-1.8. In one line of breast cancer cells resistant to SM-164 as a single agent, SM-164 radiosensitization was mediated by intrinsic apoptosis pathway through activation of caspases-9 and -3. In a line of breast cancer cells sensitive to SM-164 as a single agent, SM-164 radiosensitization was mediated by both extrinsic and intrinsic apoptosis pathways through activation of caspases-9, -8, and -3. Consistently, blockage of caspase activation, through siRNA knockdown or treatment with a pan-caspase inhibitor z-VAD-fmk, inhibited apoptosis and abrogated SM-164 radiosensitization. Our study demonstrates that IAPs are valid radiosensitizing targets in breast cancer cells and SM-164 could be further developed as a novel class of radiosensitizers for the treatment of radioresistant breast cancer.
Collapse
|
248
|
Li X, Wang J, Shi Y. Structural mechanisms of DIAP1 auto-inhibition and DIAP1-mediated inhibition of drICE. Nat Commun 2011; 2:408. [DOI: 10.1038/ncomms1418] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 06/30/2011] [Indexed: 01/24/2023] Open
|
249
|
Cheng J, Zhu Y, He S, Lu Y, Chen J, Han B, Petrillo M, Wrzeszczynski KO, Yang S, Dai P, Zhai S, Han D, Zhang MQ, Li W, Liu X, Li H, Chen ZY, Yuan H. Functional mutation of SMAC/DIABLO, encoding a mitochondrial proapoptotic protein, causes human progressive hearing loss DFNA64. Am J Hum Genet 2011; 89:56-66. [PMID: 21722859 DOI: 10.1016/j.ajhg.2011.05.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/03/2011] [Accepted: 05/25/2011] [Indexed: 11/29/2022] Open
Abstract
SMAC/DIABLO is a mitochondrial proapoptotic protein that is released from mitochondria during apoptosis and counters the inhibitory activities of inhibitor of apoptosis proteins, IAPs. By linkage analysis and candidate screening, we identified a heterozygous SMAC/DIABLO mutation, c.377C>T (p.Ser126Leu, refers to p.Ser71Leu in the mature protein) in a six-generation Chinese kindred characterized by dominant progressive nonsyndromic hearing loss, designated as DFNA64. SMAC/DIABLO is highly expressed in human embryonic ears and is enriched in the developing mouse inner-ear hair cells, suggesting it has a role in the development and homeostasis of hair cells. We used a functional study to demonstrate that the SMAC/DIABLO(S71L) mutant, while retaining the proapoptotic function, triggers significant degradation of both wild-type and mutant SMAC/DIABLO and renders host mitochondria susceptible to calcium-induced loss of the membrane potential. Our work identifies DFNA64 as the human genetic disorder associated with SMAC/DIABLO malfunction and suggests that mutant SMAC/DIABLO(S71L) might cause mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jing Cheng
- Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Targeting inhibitor of apoptosis proteins for therapeutic intervention. Future Med Chem 2011; 1:1509-25. [PMID: 21426063 DOI: 10.4155/fmc.09.116] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The inhibitors of apoptosis (IAP) proteins have emerged over the last decade as important targets for therapeutic intervention in human malignancies. Overexpression of IAPs has been implicated in cell survival and resistance against stress-induced apoptosis brought on by radiation and/or chemotherapeutics (currently the standard-of-care in a variety of different cancer diseases). In addition, evasion from death receptor-mediated apoptosis and regulation of NF-κB pathways and cell division have also been associated with IAP proteins. Efforts to target IAP proteins in tumors have focused mainly on designing small molecules that mimic the IAP-binding motif of the endogenous IAP antagonist, second mitochondrial activator of caspases. In addition, several other IAP-targeting strategies, including antisense oligonucleotides and transcriptional repression, have also been initiated, with the hope of providing therapeutic benefit to cancer patients.
Collapse
|