1
|
Sperrhacke M, Leitzke S, Ahrens B, Reiss K. Breakdown of Phospholipid Asymmetry Triggers ADAM17-Mediated Rescue Events in Cells Undergoing Apoptosis. MEMBRANES 2023; 13:720. [PMID: 37623781 PMCID: PMC10456294 DOI: 10.3390/membranes13080720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023]
Abstract
ADAM17, a prominent member of the "Disintegrin and Metalloproteinase" (ADAM) family, controls vital cellular functions through the cleavage of transmembrane substrates, including epidermal growth factor receptor (EGFR) ligands such as transforming growth factor (TGF)-alpha and Epiregulin (EREG). Several ADAM17 substrates are relevant to oncogenesis and tumor growth. We have presented evidence that surface exposure of phosphatidylserine (PS) is pivotal for ADAM17 to exert sheddase activity. The scramblase Xkr8 is instrumental for calcium-independent exposure of PS in apoptotic cells. Xkr8 can be dually activated by caspase-3 and by kinases. In this investigation, we examined whether Xkr8 would modulate ADAM17 activity under apoptotic and non-apoptotic conditions. Overexpression of Xkr8 in HEK293T cells led to significantly increased caspase-dependent as well as PMA-induced release of EREG and TGF-alpha. Conversely, siRNA-mediated downregulation of Xkr8 in colorectal Caco-2 cancer cells led to decreased PS externalization upon induction of apoptosis, which was accompanied by reduced shedding of endogenously expressed EREG and reduced cell survival. We conclude that Xkr8 shares with conventional scramblases the propensity to upmodulate the ADAM-sheddase function. Liberation of growth factors could serve a rescue function in cells on the pathway to apoptotic death.
Collapse
Affiliation(s)
| | | | | | - Karina Reiss
- Department of Dermatology, University of Kiel, 24105 Kiel, Germany (B.A.)
| |
Collapse
|
2
|
Mármol I, Montanel-Perez S, Royo JC, Gimeno MC, Villacampa MD, Rodríguez-Yoldi MJ, Cerrada E. Gold(I) and Silver(I) Complexes with 2-Anilinopyridine-Based Heterocycles as Multitarget Drugs against Colon Cancer. Inorg Chem 2020; 59:17732-17745. [DOI: 10.1021/acs.inorgchem.0c02922] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Inés Mármol
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
- Departamento de Farmacología y Fisiología, Unidad de Fisiología. and CIBERobn, IIS Aragón, IA2, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Sara Montanel-Perez
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
| | - José Carlos Royo
- Departamento de Farmacología y Fisiología, Unidad de Fisiología. and CIBERobn, IIS Aragón, IA2, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - M. Concepción Gimeno
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
| | - M. Dolores Villacampa
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
| | - M. Jesús Rodríguez-Yoldi
- Departamento de Farmacología y Fisiología, Unidad de Fisiología. and CIBERobn, IIS Aragón, IA2, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Elena Cerrada
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
| |
Collapse
|
3
|
Muraki M. Sensitization to cell death induced by soluble Fas ligand and agonistic antibodies with exogenous agents: A review. AIMS MEDICAL SCIENCE 2020. [DOI: 10.3934/medsci.2020011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
4
|
Yagolovich AV, Artykov AA, Dolgikh DA, Kirpichnikov MP, Gasparian ME. A New Efficient Method for Production of Recombinant Antitumor Cytokine TRAIL and Its Receptor-Selective Variant DR5-B. BIOCHEMISTRY (MOSCOW) 2019; 84:627-636. [PMID: 31238862 DOI: 10.1134/s0006297919060051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The cytokine TRAIL induces apoptosis in tumor cells of various origin without affecting normal cells. Clinical trials of TRAIL-receptor (DR4 and DR5) agonists (recombinant TRAIL or death receptors antibodies) have largely failed because most human tumors were resistant to them. Currently, a second generation of agents targeted at TRAIL-R with increased efficiency has been developed. To this end, we have developed DR5-B, a variant of TRAIL selectively interacting with DR5. We have developed a new efficient method for production of TRAIL and DR5-B using expression of these proteins in Escherichia coli strain SHuffle B. The proteins were isolated from the cytoplasmic fraction of cells and purified to a high degree of homogeneity using metal-affinity and ion-exchange chromatography. The protein yield was 211 and 173 mg from one liter of cell culture for DR5-B and TRAIL, respectively, which significantly exceeded the results obtained by other methods. DR5-B killed tumor cells of different origin more efficiently and rapidly compared with TRAIL. The resulting preparations can be used for the study of TRAIL signaling pathways and in preclinical and clinical trials as antitumor agents.
Collapse
Affiliation(s)
- A V Yagolovich
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.,Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - A A Artykov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.,Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - D A Dolgikh
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.,Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - M P Kirpichnikov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.,Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - M E Gasparian
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.
| |
Collapse
|
5
|
Bohlul E, Hasanlou F, Taromchi AH, Nadri S. TRAIL-expressing recombinant Lactococcus lactis induces apoptosis in human colon adenocarcinoma SW480 and HCT116 cells. J Appl Microbiol 2019; 126:1558-1567. [PMID: 30815963 DOI: 10.1111/jam.14237] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/31/2019] [Accepted: 02/22/2019] [Indexed: 12/21/2022]
Abstract
AIMS We investigated the ability of Lactococcus lactis, a species generally regarded as safe, to express tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) protein. The expressed protein was either cell wall anchored or secreted, and it was assessed whether this could induce apoptosis in human colon adenocarcinoma cell lines SW480 and HCT116. METHODS AND RESULTS Constructs were designed to produce either secreted or cell wall-anchored forms of human TRAIL cloned into pNZ7021 expression vector. The expression by L. lactis was confirmed by Western blotting and immunofluorescence. Induction of cell death was evaluated by coculturing transformants producing either form of TRAIL protein with the two cell lines followed by MTT assay. Gene expression of apoptosis genes, Bax and Bcl2, was assessed by qPCR. The viability of SW480 and HCT116 cells treated with recombinant L. lactis was significantly reduced. A significant change was observed in the ratio of Bax/Bcl2 expression in HCT116 cells only following treatment with the supernatant of recombinant L. lactis containing secreted TRAIL. CONCLUSION Recombinant L. lactis producing TRAIL protein can induce apoptosis in human colon adenocarcinoma cell lines SW480 and HCT116. SIGNIFICANCE AND IMPACT OF THE STUDY The use of recombinant probiotics that produce anticancer compounds is a promising option for combating cancer cells.
Collapse
Affiliation(s)
- E Bohlul
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - F Hasanlou
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - A H Taromchi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - S Nadri
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
6
|
Lee MW, Kim DS, Kim HR, Park HJ, Lee JW, Sung KW, Koo HH, Yoo KH. Inhibition of N-myc expression sensitizes human neuroblastoma IMR-32 cells expressing caspase-8 to TRAIL. Cell Prolif 2019; 52:e12577. [PMID: 30724400 PMCID: PMC6536445 DOI: 10.1111/cpr.12577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/21/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022] Open
Abstract
Objectives This study aims to explore the roles of N‐myc and caspase‐8 in TRAIL‐resistant IMR‐32 cells which exhibit MYCN oncogene amplification and lack caspase‐8 expression. Materials and methods We established N‐myc–downregulated IMR‐32 cells using shRNA lentiviral particles targeting N‐myc and examined the effect the N‐myc inhibition on TRAIL susceptibility in human neuroblastoma IMR‐32 cells expressing caspase‐8. Results Cisplatin treatment in IMR‐32 cells increased the expression of death receptor 5 (DR5; TRAIL‐R2), but not other receptors, via downregulation of NF‐κB activity. However, the cisplatin‐mediated increase in DR5 failed to induce cell death following TRAIL treatment. Furthermore, interferon (IFN)‐γ pretreatment increased caspase‐8 expression in IMR‐32 cells, but cisplatin failed to trigger TRAIL cytotoxicity. We downregulated N‐myc expression in IMR‐32 cells using N‐myc–targeting shRNA. These cells showed decreased growth rate and Bcl‐2 expression accompanied by a mild collapse in the mitochondrial membrane potential as compared with those treated with scrambled shRNA. TRAIL treatment in N‐myc–negative cells expressing caspase‐8 following IFN‐γ treatment significantly triggered apoptotic cell death. Concurrent treatment with cisplatin enhanced TRAIL‐mediated cytotoxicity, which was abrogated by an additional pretreatment with DR5:Fc chimera protein. Conclusions N‐myc and caspase‐8 expressions are involved in TRAIL susceptibility in IMR‐32 cells, and the combination of treatment with cisplatin and TRAIL may serve as a promising strategy for the development of therapeutics against neuroblastoma that is controlled by N‐myc and caspase‐8 expression.
Collapse
Affiliation(s)
- Myoung Woo Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dae Seong Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Ryung Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Jin Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
7
|
Gouveia M, Figueira J, Jardim MG, Castro R, Tomás H, Rissanen K, Rodrigues J. Poly(alkylidenimine) Dendrimers Functionalized with the Organometallic Moiety [Ru(η⁵-C₅H₅)(PPh₃)₂]⁺ as Promising Drugs Against Cisplatin-Resistant Cancer Cells and Human Mesenchymal Stem Cells. Molecules 2018; 23:1471. [PMID: 29914219 PMCID: PMC6100097 DOI: 10.3390/molecules23061471] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/11/2018] [Accepted: 06/12/2018] [Indexed: 02/05/2023] Open
Abstract
Here and for the first time, we show that the organometallic compound [Ru(η⁵-C₅H₅)(PPh₃)₂Cl] (RuCp) has potential to be used as a metallodrug in anticancer therapy, and further present a new approach for the cellular delivery of the [Ru(η⁵-C₅H₅)(PPh₃)₂]⁺ fragment via coordination on the periphery of low-generation poly(alkylidenimine) dendrimers through nitrile terminal groups. Importantly, both the RuCp and the dendrimers functionalized with [Ru(η⁵-C₅H₅)(PPh₃)₂]⁺ fragments present remarkable toxicity towards a wide set of cancer cells (Caco-2, MCF-7, CAL-72, and A2780 cells), including cisplatin-resistant human ovarian carcinoma cell lines (A2780cisR cells). Also, RuCp and the prepared metallodendrimers are active against human mesenchymal stem cells (hMSCs), which are often found in the tumor microenvironment where they seem to play a role in tumor progression and drug resistance.
Collapse
Affiliation(s)
- Marisol Gouveia
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390 Funchal, Portugal.
| | - João Figueira
- Department of Chemistry, ScilifeLab, Umeå University, KBC-Building, Linnaeus väg 6, 90736 Umeå, Sweden.
| | - Manuel G Jardim
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390 Funchal, Portugal.
| | - Rita Castro
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390 Funchal, Portugal.
| | - Helena Tomás
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390 Funchal, Portugal.
| | - Kari Rissanen
- Department of Chemistry, University of Jyvaskyla, P.O. Box. 35, FI-40014 Jyväskylä, Finland.
| | - João Rodrigues
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390 Funchal, Portugal.
- School of Materials Science and Engineering/Center for Nano Energy Materials, Northwestern Polytechnical University, Xi'an 710072, China.
| |
Collapse
|
8
|
Gouveia M, Figueira J, Jardim M, Castro R, Tomás H, Rissanen K, Rodrigues J. Poly(alkylidenimine) Dendrimers Functionalized with the Organometallic Moiety [Ru(η5-C5H5)(PPh3)2]+ as Promising Drugs Against Cisplatin-Resistant Cancer Cells and Human Mesenchymal Stem Cells. Molecules 2018. [DOI: https://doi.org/10.3390/molecules23061471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
9
|
Lin CF, Lin CM, Lee KY, Wu SY, Feng PH, Chen KY, Chuang HC, Chen CL, Wang YC, Tseng PC, Tsai TT. Escape from IFN-γ-dependent immunosurveillance in tumorigenesis. J Biomed Sci 2017; 24:10. [PMID: 28143527 PMCID: PMC5286687 DOI: 10.1186/s12929-017-0317-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/19/2017] [Indexed: 12/24/2022] Open
Abstract
Immune interferon (IFN), also known as IFN-γ, promotes not only immunomodulation but also antimicrobial and anticancer activity. After IFN-γ binds to the complex of IFN-γ receptor (IFNGR) 1-IFNGR2 and subsequently activates its downstream signaling pathways, IFN-γ immediately causes transcriptional stimulation of a variety of genes that are principally involved in its biological activities. Regarding IFN-γ-dependent immunosurveillance, IFN-γ can directly suppress tumorigenesis and infection and/or can modulate the immunological status in both cancer cells and infected cells. Regarding the anticancer effects of IFN-γ, cancer cells develop strategies to escape from IFN-γ-dependent cancer immunosurveillance. Immune evasion, including the recruitment of immunosuppressive cells, secretion of immunosuppressive factors, and suppression of cytotoxic T lymphocyte responses, is speculated to be elicited by the oncogenic microenvironment. All of these events effectively downregulate IFN-γ-expressing cells and IFN-γ production. In addition to these extrinsic pathways, cancer cells may develop cellular tolerance that manifests as hyporesponsiveness to IFN-γ stimulation. This review discusses the potential escape mechanisms from IFN-γ-dependent immunosurveillance in tumorigenesis.
Collapse
Affiliation(s)
- Chiou-Feng Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan. .,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.
| | - Chih-Ming Lin
- Department of Thoracic Surgery, Lotung Poh-Ai Hospital, Yilan, 265, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 110, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Szu-Yuan Wu
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.,Department of Radiation Oncology, Wan Fang Hospital, Taipei Medical University, Taipei Medical University, Taipei, 110, Taiwan.,Department of Biotechnology, Hung Kuang University, Taichung, 433, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 110, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 110, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Chia-Ling Chen
- Translational Research Center, Taipei Medical University, Taipei, 110, Taiwan
| | - Yu-Chih Wang
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Po-Chun Tseng
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Tsung-Ting Tsai
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| |
Collapse
|
10
|
Benard A, Janssen CM, van den Elsen PJ, van Eggermond MCJA, Hoon DSB, van de Velde CJH, Kuppen PJK. Chromatin status of apoptosis genes correlates with sensitivity to chemo-, immune- and radiation therapy in colorectal cancer cell lines. Apoptosis 2015; 19:1769-78. [PMID: 25292014 DOI: 10.1007/s10495-014-1042-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The apoptosis pathway of programmed cell death is frequently deregulated in cancer. An intact apoptosis pathway is required for proper response to anti-cancer treatment. We investigated the chromatin status of key apoptosis genes in the apoptosis pathway in colorectal cancer cell lines in relation to apoptosis induced by chemo-, immune- or radiation therapy. Using chromatin immunoprecipitation (ChIP), we measured the presence of transcription-activating histone modifications H3Ac and H3K4me3 and silencing modifications H3K9me3 and H3K27me3 at the gene promoter regions of key apoptosis genes Bax, Bcl2, Caspase-9, Fas (CD95) and p53. Cell lines DLD1, SW620, Colo320, Caco2, Lovo and HT29 were treated with cisplatin, anti-Fas or radiation. The apoptotic response was measured by flow cytometry using propidium iodide and annexin V-FITC. The chromatin status of the apoptosis genes reflected the activation status of the intrinsic (Bax, Bcl2, Caspase-9 and p53) and extrinsic (Fas) pathways. An active intrinsic apoptotic pathway corresponded to sensitivity to cisplatin and radiation treatment of cell lines DLD1, SW620 and Colo320. An active Fas promoter corresponded to an active extrinsic apoptotic pathway in cell line DLD1. mRNA expression data correlated with the chromatin status of the apoptosis genes as measured by ChIP. In conclusion, the results presented in this study indicate that the balance between activating and silencing histone modifications, reflecting the chromatin status of apoptosis genes, can be used to predict the response of tumor cells to different anti-cancer therapies and could provide a novel target to sensitize tumors to obtain adequate treatment responses.
Collapse
Affiliation(s)
- Anne Benard
- Department of Surgery, K6-R, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
11
|
Möller Y, Siegemund M, Beyes S, Herr R, Lecis D, Delia D, Kontermann R, Brummer T, Pfizenmaier K, Olayioye MA. EGFR-targeted TRAIL and a Smac mimetic synergize to overcome apoptosis resistance in KRAS mutant colorectal cancer cells. PLoS One 2014; 9:e107165. [PMID: 25198428 PMCID: PMC4157814 DOI: 10.1371/journal.pone.0107165] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/04/2014] [Indexed: 11/18/2022] Open
Abstract
TRAIL is a death receptor ligand that induces cell death preferentially in tumor cells. Recombinant soluble TRAIL, however, performs poorly as an anti-cancer therapeutic because oligomerization is required for potent biological activity. We previously generated a diabody format of tumor-targeted TRAIL termed DbαEGFR-scTRAIL, comprising single-stranded TRAIL molecules (scTRAIL) and the variable domains of a humanized variant of the EGFR blocking antibody Cetuximab. Here we define the bioactivity of DbαEGFR-scTRAIL with regard to both EGFR inhibition and TRAIL receptor activation in 3D cultures of Caco-2 colorectal cancer cells, which express wild-type K-Ras. Compared with conventional 2D cultures, Caco-2 cells displayed strongly enhanced sensitivity toward DbαEGFR-scTRAIL in these 3D cultures. We show that the antibody moiety of DbαEGFR-scTRAIL not only efficiently competed with ligand-induced EGFR function, but also determined the apoptotic response by specifically directing DbαEGFR-scTRAIL to EGFR-positive cells. To address how aberrantly activated K-Ras, which leads to Cetuximab resistance, affects DbαEGFR-scTRAIL sensitivity, we generated stable Caco-2tet cells inducibly expressing oncogenic K-RasG12V. In the presence of doxycycline, these cells showed increased resistance to DbαEGFR-scTRAIL, associated with the elevated expression of the anti-apoptotic proteins cIAP2, Bcl-xL and FlipS. Co-treatment of cells with the Smac mimetic SM83 restored the DbαEGFR-scTRAIL-induced apoptotic response. Importantly, this synergy between DbαEGFR-scTRAIL and SM83 also translated to 3D cultures of oncogenic K-Ras expressing HCT-116 and LoVo colorectal cancer cells. Our findings thus support the notion that DbαEGFR-scTRAIL therapy in combination with apoptosis-sensitizing agents may be promising for the treatment of EGFR-positive colorectal cancers, independently of their KRAS status.
Collapse
Affiliation(s)
- Yvonne Möller
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Martin Siegemund
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Sven Beyes
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Ricarda Herr
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Daniele Lecis
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Domenico Delia
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Roland Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Centre for Biological Signalling Studies BIOSS, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Monilola A. Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- * E-mail:
| |
Collapse
|
12
|
Tomihara K, Fuse H, Heshiki W, Takei R, Zhang B, Arai N, Nakamori K, Noguchi M. Gemcitabine chemotherapy induces phenotypic alterations of tumor cells that facilitate antitumor T cell responses in a mouse model of oral cancer. Oral Oncol 2014; 50:457-67. [DOI: 10.1016/j.oraloncology.2014.01.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/18/2014] [Accepted: 01/23/2014] [Indexed: 11/24/2022]
|
13
|
Gallouet AS, Travert M, Bresson-Bepoldin L, Guilloton F, Pangault C, Caulet-Maugendre S, Lamy T, Tarte K, Guillaudeux T. COX-2-independent effects of celecoxib sensitize lymphoma B cells to TRAIL-mediated apoptosis. Clin Cancer Res 2014; 20:2663-73. [PMID: 24637636 DOI: 10.1158/1078-0432.ccr-13-2305] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Despite therapeutic advances, non-Hodgkin lymphomas (NHL) remain incurable. They form a group of neoplasms strongly dependent on their inflammatory microenvironment, which plays an important supportive role in tumor B-cell survival and in the resistance to antitumor immune response. New therapies must consider both tumor cells and their surrounding microenvironment EXPERIMENTAL DESIGN Stromal cells, derived from bone marrow or lymph nodes, and B cells from follicular lymphoma patients were cocultured or cultured alone with celecoxib treatment, a nonsteroidal anti-inflammatory drug, and/or TRAIL, a promising cytotoxic molecule for cancer therapy. RESULTS In this study, we show that follicular lymphoma stromal cells produce large amounts of PGE2. This production is abrogated after celecoxib treatment, targeting the COX-2 isoenzyme involved in PGE2 synthesis. Furthermore, we demonstrate that celecoxib increases apoptosis in NHL B-cell lines and in primary follicular lymphoma B cells cocultured with stromal cells, but independently of the PGE2/COX-2 axis. Finally, celecoxib increases the apoptotic activity of TRAIL. We provide evidence that celecoxib affects proliferation and sensitizes NHL B-cell lines to apoptosis through COX-2-independent effects by slowing down the cell cycle and decreasing the expression of survival proteins, such as Mcl-1. CONCLUSIONS These data suggest new potent strategies for NHL therapy combining drugs targeting both tumor B cells and survival signals provided by the tumor microenvironment.
Collapse
Affiliation(s)
- Anne-Sophie Gallouet
- Authors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, France
| | - Marion Travert
- Authors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, France
| | - Laurence Bresson-Bepoldin
- Authors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, France
| | - Fabien Guilloton
- Authors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, France
| | - Céline Pangault
- Authors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, France
| | - Sylvie Caulet-Maugendre
- Authors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, France
| | - Thierry Lamy
- Authors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, France
| | - Karin Tarte
- Authors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, France
| | - Thierry Guillaudeux
- Authors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, FranceAuthors' Affiliations: INSERM, UMR 917; Université Rennes 1; EFS Bretagne; Centre Hospitalier Universitaire (CHU), Service d'Hématologie; Biosit, SFR Biologie-Santé, Rennes; INSERM, U955, Créteil; and INSERM, U916, Bordeaux, France
| |
Collapse
|
14
|
Jalving M, Heijink DM, Koornstra JJ, Boersma-van Ek W, Zwart N, Wesseling J, Sluiter WJ, de Vries EGE, Kleibeuker JH, de Jong S. Regulation of TRAIL receptor expression by β-catenin in colorectal tumours. Carcinogenesis 2013; 35:1092-9. [PMID: 24379239 DOI: 10.1093/carcin/bgt484] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Tumour-necrosis-factor-related apoptosis-inducing ligand (TRAIL) is being investigated as a targeted cancer therapeutic and the expression of its pro-apoptotic receptors, DR4 and DR5, increases during colorectal carcinogenesis. This study investigated the role of β-catenin in the regulation of these receptors. In human colorectal adenoma and carcinoma cell lines, downregulation of β-catenin resulted in lower total DR4 and DR5 protein levels. Similarly, cell membrane expression of DR4 and DR5 was reduced after downregulation of β-catenin in colon carcinoma cells, whereas induction of β-catenin in HeLa cells led to increased cell membrane expression of DR4 and DR5. Downregulation of β-catenin decreased the recombinant human TRAIL sensitivity of human colon carcinoma cells. Activation of the transcription factor T-cell factor-4 (TCF-4) is an important function of β-catenin. Dominant-negative TCF-4 overexpression, however, did not significantly affect TRAIL receptor expression or recombinant human TRAIL sensitivity. Human colorectal adenomas (N = 158) with aberrant (cytoplasmic and nuclear) β-catenin expression had a higher percentage of immunohistochemical DR4 and DR5 staining per tumour (mean: 73 and 88%, respectively) than those with membranous β-catenin staining only (mean: 50 and 70%, respectively, P < 0.01 for both). Furthermore, aberrant β-catenin staining co-localized with DR4 and DR5 expression in 92% of adenomas. In 53 human colorectal carcinomas, aberrant β-catenin expression was present in most cases and DR4/5 expression was largely homogenous. Similarly, in adenomas from APC(min) mice, cytoplasmic β-catenin staining co-localized with staining for the murine TRAIL death receptor. In conclusion, the gradual increase in TRAIL receptor expression during colorectal carcinogenesis is at least partially mediated through increased β-catenin expression, independently of TCF-4-signalling.
Collapse
|
15
|
Flusberg DA, Sorger PK. Modulating cell-to-cell variability and sensitivity to death ligands by co-drugging. Phys Biol 2013; 10:035002. [PMID: 23735516 DOI: 10.1088/1478-3975/10/3/035002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) holds promise as an anti-cancer therapeutic but efficiently induces apoptosis in only a subset of tumor cell lines. Moreover, even in clonal populations of responsive lines, only a fraction of cells dies in response to TRAIL and individual cells exhibit cell-to-cell variability in the timing of cell death. Fractional killing in these cell populations appears to arise not from genetic differences among cells but rather from differences in gene expression states, fluctuations in protein levels and the extent to which TRAIL-induced death or survival pathways become activated. In this study, we ask how cell-to-cell variability manifests in cell types with different sensitivities to TRAIL, as well as how it changes when cells are exposed to combinations of drugs. We show that individual cells that survive treatment with TRAIL can regenerate the sensitivity and death-time distribution of the parental population, demonstrating that fractional killing is a stable property of cell populations. We also show that cell-to-cell variability in the timing and probability of apoptosis in response to treatment can be tuned using combinations of drugs that together increase apoptotic sensitivity compared to treatment with one drug alone. In the case of TRAIL, modulation of cell-to-cell variability by co-drugging appears to involve a reduction in the threshold for mitochondrial outer membrane permeabilization.
Collapse
Affiliation(s)
- Deborah A Flusberg
- Center for Cell Decision Processes, Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
| | | |
Collapse
|
16
|
Pennarun B, Kleibeuker JH, Boersma-van Ek W, Kruyt FAE, Hollema H, de Vries EGE, de Jong S. Targeting FLIP and Mcl-1 using a combination of aspirin and sorafenib sensitizes colon cancer cells to TRAIL. J Pathol 2013; 229:410-21. [PMID: 23132258 DOI: 10.1002/path.4138] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 10/15/2012] [Accepted: 10/19/2012] [Indexed: 12/16/2022]
Abstract
The multikinase inhibitor sorafenib is highly effective against certain types of cancer in the clinic and prevents colon cancer cell proliferation in vitro. Non-steroidal anti-inflammatory drugs, such as acetylsalicylic acid (aspirin), have shown activity against colon cancer cells. The aims of this study were to determine whether the combination of aspirin with sorafenib has enhanced anti-proliferative effects and increases recombinant human tumour necrosis factor-related apoptosis-inducing ligand (rhTRAIL)-induced apoptosis in the human SW948, Lovo, Colo205, Colo320, Caco-2 and HCT116 colon cancer cell lines. In four cell lines, aspirin strongly stimulated the anti-proliferative effects of sorafenib (∼four-fold enhancement) by inducing cell cycle arrest. Furthermore, combining low doses of aspirin (≤ 5 mm) and sorafenib (≤ 2.5 µm) greatly sensitized TRAIL-sensitive and TRAIL-resistant colon cancer cells to rhTRAIL, much more potently than either drug combined with rhTRAIL. The increase in rhTRAIL sensitivity was due to inhibition of FLIP and Mcl-1 protein expression following aspirin and sorafenib co-treatment, as confirmed by knock-down studies. Next, the clinical relevance of targeting FLIP and Mcl-1 in colon cancer was examined. Using immunohistochemistry, we found that Mcl-1 expression was significantly increased in colon adenoma and carcinoma patient material compared to healthy colonic epithelium, similar to the enhanced FLIP expression we recently observed in colon cancer. These results underscore the potential of combining low doses of aspirin with sorafenib to inhibit proliferation and target the anti-apoptotic proteins FLIP and Mcl-1 in colon cancer cells.
Collapse
Affiliation(s)
- Bodvael Pennarun
- Department of Medical Oncology, University of Groningen, University Medical Centre Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
17
|
Song IS, Jun SY, Na HJ, Kim HT, Jung SY, Ha GH, Park YH, Long LZ, Yu DY, Kim JM, Kim JH, Ko JH, Kim CH, Kim NS. Inhibition of MKK7-JNK by the TOR signaling pathway regulator-like protein contributes to resistance of HCC cells to TRAIL-induced apoptosis. Gastroenterology 2012; 143:1341-1351. [PMID: 22841785 DOI: 10.1053/j.gastro.2012.07.103] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 06/06/2012] [Accepted: 07/19/2012] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS The TOR signaling pathway regulator-like (TIPRL) protein, the mammalian ortholog of yeast TIP41, was identified in an expression profiling screen for factors that regulate human liver carcinogenesis. We investigated the role of human TIPRL protein in hepatocellular carcinoma (HCC). METHODS We measured the level of TIPRL in HCC and adjacent nontumor tissues from patients. We used small interfering RNAs and zebrafish to study the function of TIPRL. We used annexin V propidium iodide staining and immunoblot analyses to measure apoptosis and activation of apoptotic signaling pathways. We used confocal microscopy, coimmunoprecipitation, and glutathione-S transferase pull-down analyses to determine interactions among mitogen-activated protein kinase kinase 7 (MKK7 or MAP2K7), TIPRL, and the protein phosphatase type 2A (PP2Ac). We studied the effects of TIPRL in tumor xenografts in mice. RESULTS Levels of TIPRL were higher in HCC tissues and cell lines than nontumor tissues and primary hepatocytes. Knockdown of tiprl expression in zebrafish led to large amounts of apoptosis throughout the embryos. Incubation of HCC cells, but not primary human hepatocytes, with small interfering RNA against TIPRL (siTIPRL) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) caused prolonged activation (phosphorylation) of MKK7 and c-Jun N-terminal kinase (JNK) and led to apoptosis, indicated by cleavage of procaspase-8,-3 and of poly-(adenosine diphosphate-ribose) polymerase. TIPRL bound to MKK7 and PP2Ac and promoted the interaction between MKK7 and PP2Ac. In mice, injection of HCC xenograft tumors with siTIPRL and TRAIL led to tumor apoptosis and regression. CONCLUSIONS TIPRL is highly up-regulated in human HCC samples and cell lines, compared with noncancerous liver tissues. TIPRL prevents prolonged activation of MKK7 and JNK and TRAIL-induced apoptosis by mediating the interaction between MKK7 and PP2Ac.
Collapse
Affiliation(s)
- In Sung Song
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea
| | - Soo Young Jun
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea
| | - Hee-Jun Na
- Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea; Renal Division, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Hyun-Taek Kim
- Department of Biology, Chungnam National University, Daejeon, South Korea
| | - So Young Jung
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Ga Hee Ha
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Young-Ho Park
- Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea; Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Liang Zhe Long
- Department of Pathology, School of Medicine, Chungnam National University, Daejeon, South Korea
| | - Dae-Yeul Yu
- Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea; Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Jin-Man Kim
- Department of Pathology, School of Medicine, Chungnam National University, Daejeon, South Korea
| | - Joo Heon Kim
- Department of Pathology, Eulji University School of Medicine, Daejeon, South Korea
| | - Jeong-Heon Ko
- Daejeon-KRIBB-FHCRC Research Cooperation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, South Korea.
| | - Nam-Soon Kim
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea.
| |
Collapse
|
18
|
Duiker EW, Dijkers ECF, Lambers Heerspink H, de Jong S, van der Zee AGJ, Jager PL, Kosterink JGW, de Vries EGE, Lub-de Hooge MN. Development of a radioiodinated apoptosis-inducing ligand, rhTRAIL, and a radiolabelled agonist TRAIL receptor antibody for clinical imaging studies. Br J Pharmacol 2012; 165:2203-12. [PMID: 22014269 DOI: 10.1111/j.1476-5381.2011.01718.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE The TNF-related apoptosis inducing ligand (TRAIL) induces apoptosis through activation of the death receptors, TRAIL-R1 and TRAIL-R2. Recombinant human (rh) TRAIL and the TRAIL-R1 directed monoclonal antibody mapatumumab are currently clinically evaluated as anticancer agents. The objective of this study was to develop radiopharmaceuticals targeting the TRAIL-R1, suitable for clinical use to help understand and predict clinical efficacy in patients. EXPERIMENTAL APPROACH rhTRAIL was radioiodinated with (125) I, and conjugated mapatumumab was radiolabelled with (111) In. The radiopharmaceuticals were characterized, their in vitro stability and death receptor targeting capacities were determined and in vivo biodistribution was studied in nude mice bearing human tumour xenografts with different expression of TRAIL-R1. KEY RESULTS Labelling efficiencies, radiochemical purity, stability and binding properties were optimized for the radioimmunoconjugates. In vivo biodistribution showed rapid renal clearance of [(125) I]rhTRAIL, with highest kidney activity at 15 min and almost no detectable activity after 4 h. Activity rapidly decreased in almost all organs, except for the xenografts. Radiolabelled mapatumumab showed blood clearance between 24 and 168 h and a reduced decrease in radioactivity in the high receptor expression xenograft. CONCLUSIONS AND IMPLICATIONS rhTRAIL and mapatumumab can be efficiently radiolabelled. The new radiopharmaceuticals can be used clinically to study pharmacokinetics, biodistribution and tumour targeting, which could support evaluation of the native targeted agents in phase I/II trials.
Collapse
Affiliation(s)
- E W Duiker
- Department of Medical Oncology, University of Groningen, the Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Zecevic A, Sampath D, Ewald B, Chen R, Wierda W, Plunkett W. Killing of chronic lymphocytic leukemia by the combination of fludarabine and oxaliplatin is dependent on the activity of XPF endonuclease. Clin Cancer Res 2011; 17:4731-41. [PMID: 21632856 DOI: 10.1158/1078-0432.ccr-10-2561] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE Chronic lymphocytic leukemia (CLL) resistant to fludarabine-containing treatments responds to oxaliplatin-based therapy that contains fludarabine. We postulated that a mechanism for this activity is the incorporation of fludarabine into DNA during nucleotide excision repair (NER) stimulated by oxaliplatin adducts. EXPERIMENTAL DESIGN We analyzed CLL cell viability, DNA damage, and signaling pathways in response to treatment by fludarabine, oxaliplatin, or the combination. The dependency of the combination on oxaliplatin-induced DNA repair was investigated using siRNA in CLL cells or cell line models of NER deficiency. RESULTS Synergistic apoptotic killing was observed in CLL cells after exposure to the combination in vitro. Oxaliplatin induced DNA synthesis in CLL cells, which was inhibited by fludarabine and was eliminated by knockdown of XPF, the NER 5'-endonuclease. Wild-type Chinese hamster ovarian cells showed synergistic killing after combination treatment, whereas only additive killing was observed in cells lacking XPF. Inhibition of repair by fludarabine in CLL cells was accompanied by DNA single-strand break formation. CLL cells initiated both intrinsic and extrinsic apoptotic pathways as evidenced by the loss of mitochondrial outer membrane potential and partial inhibition of cell death upon incubation with FasL antibody. CONCLUSIONS The synergistic cell killing is caused by a mechanistic interaction that requires the initiation of XPF-dependent excision repair in response to oxaliplatin adducts, and the inhibition of that process by fludarabine incorporation into the repair patch. This combination strategy may be useful against other malignancies.
Collapse
Affiliation(s)
- Alma Zecevic
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
20
|
Pennarun B, Kleibeuker JH, Oenema T, Stegehuis JH, de Vries EGE, de Jong S. Inhibition of IGF-1R-dependent PI3K activation sensitizes colon cancer cells specifically to DR5-mediated apoptosis but not to rhTRAIL. Cell Oncol (Dordr) 2011; 34:245-59. [PMID: 21538027 DOI: 10.1007/s13402-011-0033-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2010] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) initiates apoptosis in tumor cells upon binding to its cognate agonistic receptors, death receptors 4 and 5 (DR4 and DR5). The activity of the insulin-like growth factor 1 (IGF-1) survival pathway is often increased in cancer, influencing both cell proliferation and apoptosis. We hypothesized that inhibiting the IGF-1 receptor (IGF-1R) using NVP-AEW541, a small molecular weight tyrosine kinase inhibitor of the IGF-1R, could increase death receptor (DR)-mediated apoptosis in colon cancer cells. METHODS The analyses were performed by caspase assay, flow cytometry, Western blotting, immunoprecipitation and fluorescent microscopy. RESULTS Preincubation with NVP-AEW541 surprisingly decreased apoptosis induced by recombinant human TRAIL (rhTRAIL) or an agonistic DR4 antibody while sensitivity to an agonistic DR5 antibody was increased. NVP-AEW541 could inhibit IGF-1-induced activation of the phosphatidylinositol 3-kinase (PI3K) pathway. The effects of the PI3K inhibitor LY294002 on TRAIL-induced apoptosis were similar to those of NVP-AEW541, further supporting a role for IGF-1R-mediated activation of PI3K. We show that PI3K inhibition enhances DR5-mediated caspase 8 processing but also lowers DR4 membrane expression and DR4-mediated caspase 8 processing. Inhibition of PI3K reduced rhTRAIL sensitivity independently of the cell line preference for either DR4- or DR5-mediated apoptosis signaling. CONCLUSIONS Our study indicates that individual effects on DR4 and DR5 apoptosis signaling should be taken into consideration when combining DR-ligands with PI3K inhibition.
Collapse
Affiliation(s)
- Bodvael Pennarun
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, the Netherlands
| | | | | | | | | | | |
Collapse
|
21
|
Luetzkendorf J, Mueller LP, Mueller T, Caysa H, Nerger K, Schmoll HJ. Growth inhibition of colorectal carcinoma by lentiviral TRAIL-transgenic human mesenchymal stem cells requires their substantial intratumoral presence. J Cell Mol Med 2011; 14:2292-304. [PMID: 19508388 PMCID: PMC3822570 DOI: 10.1111/j.1582-4934.2009.00794.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Colorectal carcinoma (CRC) constitutes a common malignancy with limited therapeutic options in metastasized stages. Mesenchymal stem cells (MSC) home to tumours and may therefore serve as a novel therapeutic tool for intratumoral delivery of antineoplastic factors. Tumour necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) which promises apoptosis induction preferentially in tumour cells represents such a factor. We generated TRAIL-MSC by transduction of human MSC with a third generation lentiviral vector system and analysed their characteristics and capacity to inhibit CRC growth. (1) TRAIL-MSC showed stable transgene expression with neither changes in the defining MSC characteristics nor signs of malignant transformation. (2) Upon direct in vitro coculture TRAIL-MSC induced apoptosis in TRAIL-sensitive CRC-cell lines (DLD-1 and HCT-15) but also in CRC-cell lines resistant to soluble TRAIL (HCT-8 and SW480). (3) In mixed subcutaneous (s.c.) xenografts TRAIL-MSC inhibited CRC-tumour growth presumably by apoptosis induction but a substantial proportion of TRAIL-MSC within the total tumour cell number was needed to yield such anti-tumour effect. (4) Systemic application of TRAIL-MSC had no effect on the growth of s.c. DLD-1 xenografts which appeared to be due to a pulmonary entrapment and low rate of tumour integration of TRAIL-MSC. Systemic TRAIL-MSC caused no toxicity in this model. (5) Wild-type MSC seemed to exert a tumour growth-supporting effect in mixed s.c. DLD-1 xenografts. These novel results support the idea that lentiviral TRAIL-transgenic human MSC may serve as vehicles for clinical tumour therapy but also highlight the need for further investigations to improve tumour integration of transgenic MSC and to clarify a potential tumour-supporting effect by MSC.
Collapse
Affiliation(s)
- Jana Luetzkendorf
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | | | | | | | | | | |
Collapse
|
22
|
van Geelen CM, Pennarun B, Le PT, de Vries EG, de Jong S. Modulation of TRAIL resistance in colon carcinoma cells: different contributions of DR4 and DR5. BMC Cancer 2011; 11:39. [PMID: 21272366 PMCID: PMC3045356 DOI: 10.1186/1471-2407-11-39] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 01/27/2011] [Indexed: 11/23/2022] Open
Abstract
Background rhTRAIL is a therapeutic agent, derived from the TRAIL cytokine, which induces apoptosis in cancer cells by activating the membrane death receptors 4 and 5 (DR4 and DR5). Here, we investigated each receptor's contribution to rhTRAIL sensitivity and rhTRAIL resistance. We assessed whether agonistic DR4 or DR5 antibodies could be used to circumvent rhTRAIL resistance, alone or in combination with various chemotherapies. Methods Our study was performed in an isogenic model comprised of the SW948 human colon carcinoma cell line and its rhTRAIL resistant sub-line SW948-TR. Effects of rhTRAIL and agonistic DR4/DR5 antibodies on cell viability were measured using MTT assays and identification of morphological changes characteristic of apoptosis, after acridine orange staining. Sensitivity to the different death receptor ligands was stimulated using pretreatment with the cytokine IFN-gamma and the proteasome inhibitor MG-132. To investigate the mechanisms underlying the changes in rhTRAIL sensitivity, alterations in expression levels of targets of interest were measured by Western blot analysis. Co-immunoprecipitation was used to determine the composition of the death-inducing signalling complex at the cell membrane. Results SW948 cells were sensitive to all three of the DR-targeting agents tested, although the agonistic DR5 antibody induced only weak caspase 8 cleavage and limited apoptosis. Surprisingly, agonistic DR4 and DR5 antibodies induced equivalent DISC formation and caspase 8 cleavage at the level of their individual receptors, suggesting impairment of further caspase 8 processing upon DR5 stimulation. SW948-TR cells were cross-resistant to all DR-targeting agents as a result of decreased caspase 8 expression levels. Caspase 8 protein expression was restored by MG-132 and IFN-gamma pretreatment, which also re-established sensitivity to rhTRAIL and agonistic DR4 antibody in SW948-TR. Surprisingly, MG-132 but not IFN-gamma could also increase DR5-mediated apoptosis in SW948-TR. Conclusions These results highlight a critical difference between DR4- and DR5-mediated apoptotic signaling modulation, with possible implications for future combinatorial regimens.
Collapse
Affiliation(s)
- Caroline Mm van Geelen
- Departments of Medical Oncology and Gastroenterology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | | | | | | |
Collapse
|
23
|
A bioinformatical and functional approach to identify novel strategies for chemoprevention of colorectal cancer. Oncogene 2011; 30:2026-36. [PMID: 21217777 DOI: 10.1038/onc.2010.578] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Comparing normal colorectal mucosa and adenomas focusing on deregulated pathways obtains insight into the biological processes of early colorectal carcinogenesis. Publicly available microarray expression data from 26 normal mucosa and 47 adenoma samples were analyzed. Biological pathways enriched in adenomas were identified with Gene Set Enrichment Analysis (GSEA). The analysis revealed 10, 11 and 16 gene sets distinguishing adenomas from normal mucosa according to Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Map Annotator and Pathway Profiler (GenMAPP) and Biocarta databases, respectively. Biological pathways known to be involved in colon carcinogenesis such as cell cycle (P=0.002) and Wnt signaling (P=0.007) were enriched in adenomas. In addition, we found enrichment of novel pathways such as retinoblastoma (Rb) pathway (P=0.002), Src pathway (P=0.004), folate biosynthesis (P=0.019) and Hedgehog signaling (P=0.037) in adenomas. Microarray results for Rb and Src pathway genes were validated by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) on mRNA isolated from an independent set of adenoma and normal colon samples. A high correlation between microarray data and qRT-PCR expression data was found. The relevance of targeting of identified pathways was shown using the Rb pathway inhibitors roscovitine and PD-0332991 and the Src pathway inhibitor dasatinib. All inhibitors used induced cell growth reduction in adenoma cells. This study shows a bioinformatical and functional approach leading to potentially new options for chemoprevention of colorectal cancer.
Collapse
|
24
|
Heijink DM, Jalving M, Oosterhuis D, Sloots IA, Koster R, Hollema H, Kleibeuker JH, Koornstra JJ, de Vries EGE, de Jong S. TNF-related apoptosis-inducing ligand cooperates with NSAIDs via activated Wnt signalling in (pre)malignant colon cells. J Pathol 2010; 223:378-89. [PMID: 21171083 DOI: 10.1002/path.2797] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2010] [Revised: 09/08/2010] [Accepted: 09/24/2010] [Indexed: 11/11/2022]
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) receptor agonistic agents and non-steroidal anti-inflammatory drugs (NSAIDs) are interesting agents for the chemoprevention and treatment of colorectal cancer. We investigated whether NSAIDs sensitize colon cancer and adenoma cell lines and ex vivo cultured human adenomas to recombinant human (rh)TRAIL. Involvement of the crucial Wnt signalling pathway in the sensitization of colon cancer cells was examined. Five colon cancer and two adenoma cell lines, human ex vivo adenomas and normal colonic epithelium were treated with aspirin or sulindac combined with rhTRAIL. Apoptosis levels, expression of intracellular proteins and TRAIL receptor membrane expression were assessed. Ls174T cells stably transfected with an inducible dominant negative TCF-4 (dnTCF-4) construct served to analyse the role of Wnt pathway activation. Both rhTRAIL-sensitive and -resistant colon cancer cell lines were strongly sensitized to rhTRAIL by aspirin (maximum enhancement ratio, 7.1). Remarkably, in adenoma cell lines sulindac enhanced rhTRAIL-induced apoptosis most effectively (maximum enhancement ratio, 2.5). Although membrane TRAIL receptor expression was not affected by NSAIDs, caspase-8 activation was enhanced by combinational treatment. Several proteins from different biological pathways were affected by NSAIDs, indicating complex mechanisms of sensitization. Elimination of TCF-4 completely blocked the sensitizing effect in colon cancer cells. In ex vivo adenomas the combination of sulindac and rhTRAIL increased apoptosis from 18.4% (sulindac) and 17.8% (rhTRAIL) to 28.0% (p = 0.003 and p = 0.005, respectively). It was concluded that NSAID-induced sensitization to rhTRAIL requires TCF-4 activity. Thus, the combination of TRAIL-receptor agonistic agents and NSAIDs is a potentially attractive treatment option for (pre)malignant tumours with constitutively active Wnt signalling, such as colorectal tumours.
Collapse
Affiliation(s)
- Dianne M Heijink
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Jani TS, DeVecchio J, Mazumdar T, Agyeman A, Houghton JA. Inhibition of NF-kappaB signaling by quinacrine is cytotoxic to human colon carcinoma cell lines and is synergistic in combination with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) or oxaliplatin. J Biol Chem 2010; 285:19162-72. [PMID: 20424169 DOI: 10.1074/jbc.m109.091645] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Colorectal cancer is the third most common malignancy in the United States. Modest advances with therapeutic approaches that include oxaliplatin (L-OHP) have brought the median survival rate to 22 months, with drug resistance remaining a significant barrier. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is undergoing clinical evaluation. Although human colon carcinomas express TRAIL receptors, they can also demonstrate TRAIL resistance. Constitutive NF-kappaB activation has been implicated in resistance to TRAIL and to cytotoxic agents. We have demonstrated constitutive NF-kappaB activation in five of six human colon carcinoma cell lines; this activation is inhibited by quinacrine. Quinacrine induced apoptosis in colon carcinomas and potentiated the cytotoxic activity of TRAIL in RKO and HT29 cells and that of L-OHP in HT29 cells. Similarly, overexpression of IkappaBalpha mutant (IkappaBalphaM) or treatment with the IKK inhibitor, BMS-345541, also sensitized these cells to TRAIL and L-OHP. Importantly, 2 h of quinacrine pretreatment resulted in decreased expression of c-FLIP and Mcl-1, which were determined to be transcriptional targets of NF-kappaB. Extended exposure for 24 h to quinacrine did not further sensitize these cells to TRAIL- or L-OHP-induced cell death; however, exposure caused the down-regulation of additional NF-kappaB-dependent survival factors. Short hairpin RNA-mediated knockdown of c-FLIP or Mcl-1 significantly sensitized these cells to TRAIL and L-OHP. Taken together, data demonstrate that NF-kappaB is constitutively active in colon cancer cell lines and NF-kappaB, and its downstream targets may constitute an important target for the development of therapeutic approaches against this disease.
Collapse
Affiliation(s)
- Tanvi S Jani
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | |
Collapse
|
26
|
Abstract
Background: Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) induces tumour cell apoptosis by binding to death receptor 4 (DR4) and DR5. DR4 and DR5 activation however can also induce inflammatory and pro-survival signalling. It is not known how these different cellular responses are regulated and what the individual role of DR4 vs DR5 is in these processes. Methods: DNA microarray study was carried out to identify genes differentially expressed after DR4 and DR5 activation. RT–PCR and western blotting was used to examine the expression of early growth response gene-1 (Egr-1) and the proteins of the TRAIL signalling pathway. The function of Egr-1 was studied by siRNA-mediated knockdown and overexpression of a dominant-negative version of Egr-1. Results: We show that the immediate early gene, Egr-1, regulates TRAIL sensitivity. Egr-1 is constitutively expressed in colon cancer cells and further induced upon activation of DR4 or DR5. Our results also show that DR4 mediates a type II, mitochondrion-dependent apoptotic pathway, whereas DR5 induces a mitochondrion-independent, type I apoptosis in HCT15 colon carcinoma cells. Egr-1 drives c-FLIP expression and the short splice variant of c-FLIP (c-FLIPS) specifically inhibits DR5 activation. Conclusion: Selective knockdown of c-FLIPS sensitises cells to DR5-induced but not DR4-induced apoptosis and Egr-1 exerts an effect as an inhibitor of the DR5-induced apoptotic pathway, possibly by regulating the expression of c-FLIPS.
Collapse
|
27
|
Dijkers EC, Kosterink JG, Rademaker AP, Perk LR, van Dongen GA, Bart J, de Jong JR, de Vries EG, Lub-de Hooge MN. Development and Characterization of Clinical-Grade 89Zr-Trastuzumab for HER2/neu ImmunoPET Imaging. J Nucl Med 2009; 50:974-81. [DOI: 10.2967/jnumed.108.060392] [Citation(s) in RCA: 261] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
28
|
Duiker EW, de Vries EG, Mahalingam D, Meersma GJ, Boersma-van Ek W, Hollema H, Lub-de Hooge MN, van Dam GM, Cool RH, Quax WJ, Samali A, van der Zee AG, de Jong S. Enhanced Antitumor Efficacy of a DR5-Specific TRAIL Variant over Recombinant Human TRAIL in a Bioluminescent Ovarian Cancer Xenograft Model. Clin Cancer Res 2009; 15:2048-57. [DOI: 10.1158/1078-0432.ccr-08-1535] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
29
|
Reis CR, van der Sloot AM, Szegezdi E, Natoni A, Tur V, Cool RH, Samali A, Serrano L, Quax WJ. Enhancement of Antitumor Properties of rhTRAIL by Affinity Increase toward Its Death Receptors. Biochemistry 2009; 48:2180-91. [DOI: 10.1021/bi801927x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Carlos R. Reis
- Department of Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Centre for Genomic Regulation, CRG-EMBL Systems Biology Unit, Dr. Aiguader 88, 08003, Barcelona, Spain, Department of Biochemistry and National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland, Triskel Therapeutics BV, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, and Institució Catalana de Recerca I Estudis Avançats (ICREA),
| | - Almer M. van der Sloot
- Department of Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Centre for Genomic Regulation, CRG-EMBL Systems Biology Unit, Dr. Aiguader 88, 08003, Barcelona, Spain, Department of Biochemistry and National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland, Triskel Therapeutics BV, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, and Institució Catalana de Recerca I Estudis Avançats (ICREA),
| | - Eva Szegezdi
- Department of Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Centre for Genomic Regulation, CRG-EMBL Systems Biology Unit, Dr. Aiguader 88, 08003, Barcelona, Spain, Department of Biochemistry and National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland, Triskel Therapeutics BV, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, and Institució Catalana de Recerca I Estudis Avançats (ICREA),
| | - Alessandro Natoni
- Department of Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Centre for Genomic Regulation, CRG-EMBL Systems Biology Unit, Dr. Aiguader 88, 08003, Barcelona, Spain, Department of Biochemistry and National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland, Triskel Therapeutics BV, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, and Institució Catalana de Recerca I Estudis Avançats (ICREA),
| | - Vicente Tur
- Department of Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Centre for Genomic Regulation, CRG-EMBL Systems Biology Unit, Dr. Aiguader 88, 08003, Barcelona, Spain, Department of Biochemistry and National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland, Triskel Therapeutics BV, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, and Institució Catalana de Recerca I Estudis Avançats (ICREA),
| | - Robbert H. Cool
- Department of Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Centre for Genomic Regulation, CRG-EMBL Systems Biology Unit, Dr. Aiguader 88, 08003, Barcelona, Spain, Department of Biochemistry and National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland, Triskel Therapeutics BV, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, and Institució Catalana de Recerca I Estudis Avançats (ICREA),
| | - Afshin Samali
- Department of Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Centre for Genomic Regulation, CRG-EMBL Systems Biology Unit, Dr. Aiguader 88, 08003, Barcelona, Spain, Department of Biochemistry and National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland, Triskel Therapeutics BV, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, and Institució Catalana de Recerca I Estudis Avançats (ICREA),
| | - Luis Serrano
- Department of Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Centre for Genomic Regulation, CRG-EMBL Systems Biology Unit, Dr. Aiguader 88, 08003, Barcelona, Spain, Department of Biochemistry and National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland, Triskel Therapeutics BV, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, and Institució Catalana de Recerca I Estudis Avançats (ICREA),
| | - Wim J. Quax
- Department of Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Centre for Genomic Regulation, CRG-EMBL Systems Biology Unit, Dr. Aiguader 88, 08003, Barcelona, Spain, Department of Biochemistry and National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland, Triskel Therapeutics BV, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, and Institució Catalana de Recerca I Estudis Avançats (ICREA),
| |
Collapse
|
30
|
Aguilera DG, Das CM, Sinnappah-Kang ND, Joyce C, Taylor PH, Wen S, Hasselblatt M, Paulus W, Fuller G, Wolff JE, Gopalakrishnan V. Reactivation of death receptor 4 (DR4) expression sensitizes medulloblastoma cell lines to TRAIL. J Neurooncol 2009; 93:303-18. [PMID: 19148581 DOI: 10.1007/s11060-008-9788-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Accepted: 12/30/2008] [Indexed: 01/21/2023]
Abstract
OBJECT Apoptosis, a key cellular response to therapeutic agents is often inactivated in tumor cells. In this study, we evaluated the expression of the tumor necrosis family of death receptors, DR4 and DR5, in medulloblastoma tumor samples and cell lines to determine if epigenetic modulation of gene expression could sensitize tumor cell lines to TRAIL-mediated apoptosis. METHODS Human medulloblastoma samples and cell lines were analyzed for DR4 and DR5 expression by quantitative PCR and immunofluorescence assays. Cell lines with downregulated expression of one or both genes were treated with the histone deacetylase inhibitor, MS-275, and the expression of DR4 and DR5 measured by quantitative PCR, Western blotting, flow cytometry and chromatin immunoprecipitation assays. Induction of apoptosis in the presence of MS-275 was evaluated by TUNEL assay and its ability to augment TRAIL-mediated cytotoxicity was determined by MTT assays, Western blotting and flow cytometry. RESULTS Compared to normal cerebellum, DR4, but not DR5 expression was consistently downregulated in medulloblastoma tumor samples and in Daoy and D283 cell lines. Interestingly, MS-275 decreased cell growth and induced apoptosis in Daoy and D283 cells. In Daoy cells, this coincided with increased histone H3 and H4 acetylation at the DR4 promoter and enhanced DR4 gene and protein expression as well as elevated Caspase-8 activity. The involvement of DR4 in the cellular response to MS-275 was further confirmed by the observation that knockdown of DR4 and FADD abrogated apoptosis. Further, addition of TRAIL to MS-275 treated cells resulted in an enhancement of apoptosis, suggesting that the upregulated death receptors were functional. CONCLUSION Our study provides an understanding of the role of DR4 in apoptosis of medulloblastoma cell lines and suggests a potential contribution of aberrant histone deacetylation to the resistance of medulloblastoma cells to therapeutic death.
Collapse
Affiliation(s)
- Dolly G Aguilera
- Department of Pediatrics, University of Texas, M.D. Anderson Cancer Center, Houston, 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Herszényi L, Farinati F, Miheller P, Tulassay Z. Chemoprevention of colorectal cancer: feasibility in everyday practice? Eur J Cancer Prev 2008; 17:502-514. [PMID: 18941372 DOI: 10.1097/cej.0b013e3282f0c080] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chemoprevention means the use of agents to prevent, delay, or reverse carcinogenesis. This review was designed to critically discuss the most promising agents in colorectal cancer (CRC) chemoprevention. Aspirin is the best studied chemopreventive agent for CRC. Optimal chemoprevention requires long-term use and high dose of aspirin that may increase the risk of gastrointestinal bleeding. Nonsteroidal anti-inflammatory drugs and selective cyclooxygenase-2 inhibitors may also be candidates for chemoprevention. The regular use of nonsteroidal anti-inflammatory drugs, however, causes adverse effects including gastrointestinal bleeding, and cyclooxygenase-2 inhibitors may increase the risk of cardiovascular events. In patients with ulcerative colitis 5-aminosalicylates reduce the risk of CRC and dysplasia. Ursodeoxycholic acid can reduce the risk of dysplasia or CRC in patients with primary sclerosing cholangitis and ulcerative colitis. Current data are insufficient to support the use of hormone replacement therapy to reduce the risk of CRC. Statins may have chemopreventive effects, but further investigation of their overall benefits in preventing CRC is warranted. Antioxidant supplements cannot prevent CRC. The usefulness of selenium, folate, calcium, and vitamin D awaits further evaluation. Chemoprevention cannot yet be accepted as standard medical practice. Use of chemopreventive agents cannot be a substitute for colorectal surveillance.
Collapse
Affiliation(s)
- László Herszényi
- 2nd Department of Medicine, Semmelweis University, Hungarian Academy of Science, Budapest, Hungary.
| | | | | | | |
Collapse
|
32
|
Trail Receptors: Targets for Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 615:127-58. [DOI: 10.1007/978-1-4020-6554-5_7] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
33
|
Li Y, Lim SC. Cadmium-induced apoptosis of hepatocytes is not associated with death receptor-related caspase-dependent pathways in the rat. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2007; 24:231-238. [PMID: 21783816 DOI: 10.1016/j.etap.2007.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Revised: 05/16/2007] [Accepted: 05/23/2007] [Indexed: 02/07/2023]
Abstract
Cadmium (Cd) is a heavy metal of considerable environmental and occupational concern. The liver is the major target organ of Cd toxicity that follows from repeated exposure to Cd. The aim of this study was to investigate the mechanism of cell death of Cd-induced hepatotoxicity in a rat model. Eighteen adult male Sprague-Dawley (SD) rats were injected daily with a dose of Cd acetate (30μM/kg body weight, subcutaneously). After 1, 2 and 7 days rats were euthanized and blood and liver tissues were sampled for analysis. Biochemical analyses of the level of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were undertaken. Histopathological and Western blot analyses for liver cellular damage and cell death were also performed. The results for the Cd-treated group of animals were compared to those from 12 control rats. The serum AST/ALT levels increased significantly 24h after CD exposure. From the Western blot analyses, activation of Bid, independent of caspase-8 was seen and Bax induced the release of cytochrome c into the cytosol from mitochondria in a dose-dependent manner. The level of Bcl-2 was decreased. Eventually, caspase-9 and caspase-3 were activated, and poly(ADP-ribose) polymerase (PARP) was cleaved in a dose-dependent manner. A histopathological analysis and DNA fragmentation test showed apoptotic cell death of the hepatocytes increased over time. These results suggest that Cd-induced liver cell apoptosis in the rat, over a period of 7 days, may not be related to the death-receptor pathway. Moreover, apoptosis is dose-dependent and associated with the decrement of Bcl-2.
Collapse
Affiliation(s)
- Yong Li
- Department of Pathology, College of Medicine, Chosun University, Seosuk-dong, Dong-gu, Gwangju 501-759, Republic of Korea
| | | |
Collapse
|
34
|
Takeda K, Stagg J, Yagita H, Okumura K, Smyth MJ. Targeting death-inducing receptors in cancer therapy. Oncogene 2007; 26:3745-57. [PMID: 17530027 DOI: 10.1038/sj.onc.1210374] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Deregulated cell death pathways may lead to the development of cancer, and induction of tumor cell apoptosis is the basis of many cancer therapies. Knowledge accumulated concerning the molecular mechanisms of apoptotic cell death has aided the development of new therapeutic strategies to treat cancer. Signals through death receptors of the tumor necrosis factor (TNF) superfamily have been well elucidated, and death receptors are now one of the most attractive therapeutic targets in cancer. In particular, DR5 and DR4, death receptors of TNF-related apoptosis-inducing ligand (TRAIL or Apo2L), are interesting targets of antibody-based therapy, since TRAIL may also bind decoy receptors that may prevent TRAIL-mediated apoptosis, whereas TRAIL ligand itself selectively induces apoptosis in cancer cells. Here, we review the potential therapeutic utility of agonistic antibodies against DR5 and DR4 and discuss the possible extension of this single-antibody-based strategy when combined with additional modalities that either synergizes to cause enhanced apoptosis or further engage the cellular immune response. Rational design of antibody-based therapies combining the induction of tumor cell apoptosis and activation of tumor-specific adaptive immunity enables promotion of distinct steps of the antitumor immune response, thereby enhancing tumor-specific lymphocytes that can eradicate TRAIL/DR5-resistant mutating, large established and heterogeneous tumors in a manner that does not require the definition of individual tumor-specific antigens.
Collapse
Affiliation(s)
- K Takeda
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
35
|
Kim H, Chaudhuri TR, Buchsbaum DJ, Wang D, Zinn KR. High-resolution single-photon emission computed tomography and X-ray computed tomography imaging of Tc-99m-labeled anti-DR5 antibody in breast tumor xenografts. Mol Cancer Ther 2007; 6:866-75. [PMID: 17363481 DOI: 10.1158/1535-7163.mct-06-0230] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A murine, apoptosis-inducing monoclonal antibody (mTRA-8) targeting human DR5 was radiolabeled with Tc-99m. The binding affinity (K(d)) and the number of DR5 receptors were measured in MD MBA-231-derived 2LMP cell lines that were "sensitive" or "resistant" to mTRA-8 killing. Single-photon emission computed tomography and X-ray computed tomography (SPECT/CT) evaluated the Tc-99m-mTRA-8 retention and distribution within xenograft tumors; biodistribution analyses confirmed the levels. Scatchard assays showed specific and high binding affinity of Tc-99m-mTRA-8 to DR5; the killing efficacy of mTRA-8 was unchanged by Tc-99m labeling. There was no significant difference between sensitive and resistant 2LMP cells for K(d) values (1.5 +/- 0.3 nmol/L = acid labile), or DR5 receptors (mean/cell = 11,000). SPECT/CT imaging analyses at 6 h after injection of Tc-99m-mTRA-8 revealed the second 1.5 mm shell from the surface of the mammary fat pad tumors (n = 5; 5,627 mm(3)) retained 12.7 +/- 1.4%ID/g, higher than the other shells, with no difference between the sensitive and resistant 2LMP tumors. Binding of Tc-99m-labeled mTRA-8 in tumor was specific; excess unlabeled mTRA-8 blocked Tc-99m-mTRA-8 retention in tumor by 45%. Retention of Tc-99m-labeled isotype antibody in tumor was consistent with the blocking study, and 30% lower. These studies show that SPECT/CT imaging provided detailed distribution information of Tc-99m-labeled mTRA-8 within breast tumor xenografts. Imaging could provide a mechanism to assess DR5 modulation when DR5 therapy is combined with chemotherapy and radiation, and thereby aid in optimizing the dosing schedule.
Collapse
Affiliation(s)
- Hyunki Kim
- Department of Radiology, Comprehensive Cancer Center, University of Alabama at Birmingham, 1808 7th Avenue South, Birmingham, AL 35294-0012, USA
| | | | | | | | | |
Collapse
|
36
|
Lippa MS, Strockbine LD, Le TT, Branstetter DG, Strathdee CA, Holland PM. Expression of anti-apoptotic factors modulates Apo2L/TRAIL resistance in colon carcinoma cells. Apoptosis 2007; 12:1465-78. [PMID: 17440816 DOI: 10.1007/s10495-007-0076-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) selectively induces apoptosis in transformed cells. Normal cells and certain tumor cells can evade Apo2L/TRAIL induced cell death, but the determinants of Apo2L/TRAIL sensitivity are poorly understood. To better understand the factors that contribute to Apo2L/TRAIL resistance, we characterized two colon carcinoma lines with pronounced differences in Apo2L/TRAIL sensitivity. Colo205 cells are highly sensitive to Apo2L/TRAIL whereas Colo320 cells are unresponsive. Components of the DISC (death inducing signaling complex) could be immunoprecipitated from both cell lines in response to Apo2L/TRAIL. Sensitizing agents including a proteasome inhibitor conferred Apo2L/TRAIL sensitivity in Colo320 cells, indicating that the apoptotic machinery was intact and functional. We specifically suppressed the expression of Bcl-2, FLIP or XIAP in Colo320 cells. Downregulation of either FLIP or XIAP but not Bcl-2 restored sensitivity of Colo320 cells to Apo2L/TRAIL. Moreover, stable knockdown of XIAP expression in Colo320 subcutaneous tumors resulted in suppression of tumor growth and sensitivity to Apo2L/TRAIL in vivo. Our results indicate that only a specific subset of anti-apoptotic proteins can confer resistance to Apo2L/TRAIL in Colo320 cells. Elucidation of the factors that contribute to Apo2L/TRAIL resistance in tumor cells may provide insight into combination therapies with Apo2L/TRAIL in a clinical setting.
Collapse
Affiliation(s)
- Mara S Lippa
- Department of Oncology, Amgen Inc., 1201 Amgen Ct. West, Seattle, WA 98119, USA
| | | | | | | | | | | |
Collapse
|
37
|
Jalving M, de Jong S, Koornstra JJ, Boersma-van Ek W, Zwart N, Wesseling J, de Vries EGE, Kleibeuker JH. TRAIL induces apoptosis in human colorectal adenoma cell lines and human colorectal adenomas. Clin Cancer Res 2007; 12:4350-6. [PMID: 16857810 DOI: 10.1158/1078-0432.ccr-05-2487] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Recombinant human (rh) tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potential new anticancer drug which can induce apoptosis in colorectal cancer cell lines. The aim of this study was to investigate whether it is possible to induce apoptosis in human adenoma cell lines and human adenomas using rhTRAIL. EXPERIMENTAL DESIGN Two human adenoma cell lines were exposed to 0.1 microg/mL of rhTRAIL for 5 hours. Apoptosis and caspase activation in cell lines were evaluated using immunocytochemistry, fluorimetric caspase assays, and Western blotting. Short-term explant cultures were established from freshly removed human adenomas (n = 38) and biopsies of normal colon epithelium (n = 15), and these were incubated for 5 hours in the presence or absence of 1 microg/mL of rhTRAIL. Apoptosis was determined in paraffin-embedded tissue using morphologic criteria and cleaved caspase-3 staining. RESULTS In the adenoma cell lines, rhTRAIL induced up to 55% apoptosis. This coincided with caspase-8 and caspase-3 activation and could be inhibited by a pan-caspase inhibitor. rhTRAIL induced caspase-dependent apoptosis in adenomas with high-grade dysplasia (n = 21) compared with the paired untreated counterparts (apoptotic index, 34 +/- 5% versus 17 +/- 2%, mean +/- SE; P = 0.002), but not in adenomas with low-grade dysplasia (n = 17) or in normal colon epithelium (n = 15). CONCLUSIONS Colorectal adenoma cell lines and adenomas with high-grade dysplasia are sensitive to rhTRAIL-induced apoptosis, whereas normal colon epithelium is not. This suggests the potential application of rhTRAIL in the treatment of adenomas with high-grade dysplasia.
Collapse
Affiliation(s)
- Mathilde Jalving
- Department of Gastroenterology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Sivaprasad U, Shankar E, Basu A. Downregulation of Bid is associated with PKCɛ-mediated TRAIL resistance. Cell Death Differ 2006; 14:851-60. [PMID: 17186022 DOI: 10.1038/sj.cdd.4402077] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent as it selectively kills tumor cells but spares normal cells. Resistance to TRAIL by tumor cells limits its therapeutic use. We have previously shown that protein kinase C-epsilon (PKCepsilon) acts as an antiapoptotic protein in MCF-7 breast cancer cells. In the present study, we have investigated the mechanism(s) by which PKCepsilon contributes to TRAIL resistance. Overexpression of PKCepsilon inhibited caspase-8 and -9 activation, release of mitochondrial cytochrome c and cell death induced by TRAIL, but did not interfere with the recruitment of caspase-8 to the death-inducing signaling complex. Knockdown/inhibition of PKCepsilon resulted in enhanced sensitivity to TRAIL. The level of Bcl-2 was increased and Bid was decreased by PKCepsilon at both the protein and mRNA level but PKCepsilon had no effect on Bax. Knockdown of Bcl-2 by siRNA reversed TRAIL resistance in PKCepsilon-overexpressing cells, whereas depletion of Bid contributed to TRAIL resistance in MCF-7 cells. A decrease in Bid content was also associated with inhibition of TRAIL-induced caspase-8 activation. Furthermore, PKCepsilon depletion or overexpression of DN-PKCepsilon was associated with a decrease in Bcl-2 protein level. Thus, our results suggest that PKCepsilon acts upstream of mitochondria and mediates TRAIL resistance via both Bcl-2 and Bid in MCF-7 cells.
Collapse
Affiliation(s)
- U Sivaprasad
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | | | | |
Collapse
|
39
|
van Geelen CM, Westra JL, de Vries EG, Boersma-van Ek W, Zwart N, Hollema H, Boezen HM, Mulder NH, Plukker JT, de Jong S, Kleibeuker JH, Koornstra JJ. Prognostic significance of tumor necrosis factor-related apoptosis-inducing ligand and its receptors in adjuvantly treated stage III colon cancer patients. J Clin Oncol 2006; 24:4998-5004. [PMID: 17075118 DOI: 10.1200/jco.2006.06.8809] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE In preclinical models, there is synergism between chemotherapy and recombinant human tumor necrosis factor (TNF) -related apoptosis-inducing ligand (TRAIL) on apoptosis induction in tumor cells. Therefore, the prognostic relevance was analyzed of the expression of TRAIL and its death receptors DR4 and DR5 on disease-free survival and overall survival in stage III colon cancer patients treated with adjuvant chemotherapy. METHODS Tissue microarrays were constructed of primary tumor tissue from 376 stage III colon cancer patients treated in a randomized adjuvant chemotherapy study (fluorouracil/levamisole v fluorouracil/levamisole/leucovorin) and stained immunohistochemically for TRAIL, DR4, and DR5. Log-rank tests and Cox proportional hazard analysis, with adjustment for treatment arm, sex, age, N stage, microsatellite instability status, and p53 mutation status, were performed. RESULTS The majority of tumors showed high expression of TRAIL (83%), DR4 (92%), and DR5 (87%). Median follow-up was 43 months. High DR4 expression was associated with worse disease-free survival (odds ratio [OR] = 2.19; 95% CI, 1.06 to 4.53; P = .03), worse overall survival (OR = 2.22; 95% CI,1.03 to 4.81; P = .04) and shorter time to recurrence (P = .02) compared with those with low DR4 expression. TRAIL or DR5 expression had no prognostic value. CONCLUSION High DR4 expression is associated with worse disease-free and overall survival in stage III adjuvant-treated colon cancer patients. Evaluation of DR4 expression in stage III colon cancer patients may identify a subset requiring more aggressive adjuvant treatment.
Collapse
Affiliation(s)
- Caroline M van Geelen
- Department of Medical Oncology, University Medical Center, University of Groningen, 9700 RB Groningen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hu G, Barnes BJ. Interferon regulatory factor-5-regulated pathways as a target for colorectal cancer therapeutics. Expert Rev Anticancer Ther 2006; 6:775-84. [PMID: 16759167 DOI: 10.1586/14737140.6.5.775] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Colorectal cancer is the second most common cause of cancer-related death. A significant obstacle to successful management of patients with colorectal cancer is intrinsic drug resistance or, in patients who initially responded to chemotherapy, acquired drug resistance. Failure in normal apoptotic pathways often contributes to resistance to anticancer drugs or radiotherapy. As a result, the identification of genes that control cell death and apoptosis has come to the forefront of cancer research, leading to new targets and novel therapeutic strategies in the treatment of colorectal cancer. To this effect, the authors have recently identified a new apoptotic signaling pathway that occurs through the transcription factor interferon regulatory factor-5. Here, the different strategies for targeting the interferon regulatory factor-5 signaling pathway in colorectal cancer are discussed. These strategies can be applied to a new generation of cytotoxic agents, as well as to novel biological compounds that are directed at inducing and/or activating interferon regulatory factor-5 or key components of this pathway.
Collapse
Affiliation(s)
- Goudong Hu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | |
Collapse
|
41
|
Martin S, Phillips DC, Szekely-Szucs K, Elghazi L, Desmots F, Houghton JA. Cyclooxygenase-2 inhibition sensitizes human colon carcinoma cells to TRAIL-induced apoptosis through clustering of DR5 and concentrating death-inducing signaling complex components into ceramide-enriched caveolae. Cancer Res 2006; 65:11447-58. [PMID: 16357153 DOI: 10.1158/0008-5472.can-05-1494] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cyclooxygenase-2 (COX-2) is up-regulated in human colon carcinomas, and its inhibition is associated with a reduction in tumorigenesis and a promotion of apoptosis. However, the mechanisms responsible for the antitumor effects of COX-2 inhibitors and how COX-2 modulates apoptotic signaling have not been clearly defined. We have shown that COX-2 inhibition sensitizes human colon carcinoma cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis by inducing clustering of the TRAIL receptor DR5 at the cell surface and the redistribution of the death-inducing signaling complex components (DR5, FADD, and procaspase-8) into cholesterol-rich and ceramide-rich domains known as caveolae. This process requires the accumulation of arachidonic acid and sequential activation of acid sphingomyelinase for the generation of ceramide within the plasma membrane outer leaflet. The current study highlights a novel mechanism to circumvent colorectal carcinoma cell resistance to TRAIL-mediated apoptosis using COX-2 inhibitors to manipulate the lipid metabolism within the plasma membrane.
Collapse
Affiliation(s)
- Sophie Martin
- Division of Molecular Therapeutics, Department of Hematology-Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | |
Collapse
|
42
|
Hougardy BMT, van der Zee AGJ, van den Heuvel FAJ, Timmer T, de Vries EGE, de Jong S. Sensitivity to Fas-mediated apoptosis in high-risk HPV-positive human cervical cancer cells: relationship with Fas, caspase-8, and Bid. Gynecol Oncol 2005; 97:353-64. [PMID: 15863130 DOI: 10.1016/j.ygyno.2005.01.036] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2004] [Revised: 01/18/2005] [Accepted: 01/31/2005] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Binding of Fas ligand or agonistic anti-Fas antibody to the death receptor Fas can activate a caspase-cascade resulting in apoptosis. In the present study, the functionality of the Fas pathway was studied in human cervical cancer cells with different HPV and p53 status. METHODS HeLa (HPV-18 positive), CaSki, and SiHa (both HPV-16 positive) contain wild-type p53, while C33A (HPV negative) expresses mutant p53. Fas cell surface expression was determined by flow cytometry. Expression of proteins involved in the apoptotic pathway was analyzed by Western blotting and apoptosis was measured by acridine orange staining of nuclear chromatin. RESULTS Despite high Fas membrane expression in the HPV-positive cells, CaSki was highly sensitive, HeLa slightly sensitive, and SiHa and C33A were resistant for agonistic anti-Fas antibody. Almost undetectable Fas membrane levels can explain the non-responsiveness of C33A for anti-Fas. Although interferon-gamma (IFNgamma) strongly and cisplatin to a lesser extend enhanced Fas membrane expression in all HPV-positive cells, sensitization to anti-Fas by IFNgamma or cisplatin was only observed in HeLa. Analysis of the Fas apoptotic pathway showed that anti-Fas treatment induced caspase-8 activation and concomitantly Bid cleavage, caspase-9 and caspase-3 activation, PARP cleavage and apoptosis in HeLa and CaSki. IFNgamma plus anti-Fas treatment, in contrast to anti-Fas alone, facilitated caspase-8 activation in HeLa and SiHa, while an increase in Bid cleavage, caspase-9 activation and apoptosis was only observed in HeLa. Apoptotic failure in SiHa (even in the presence of IFNgamma) was probably due to low caspase-8, almost undetectable Bid protein levels and therefore lack of caspase-9 activation. CONCLUSION Sensitivity to anti-Fas depends on Fas, caspase-8, and Bid protein levels in cervical cancer cells. Additionally, IFNgamma and cisplatin can increase sensitivity to anti-Fas in a subset of HPV-positive cervical cancer cell lines by upregulation of Fas and caspase-8 expression without major changes in p53 levels.
Collapse
Affiliation(s)
- Brigitte M T Hougardy
- Department of Gynecology-Oncology, University Medical Center Groningen, P.O. Box 30001, 9700 RB Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
43
|
Koornstra JJ, Jalving M, Rijcken FEM, Westra J, Zwart N, Hollema H, de Vries EGE, Hofstra RWM, Plukker JTM, de Jong S, Kleibeuker JH. Expression of tumour necrosis factor-related apoptosis-inducing ligand death receptors in sporadic and hereditary colorectal tumours: Potential targets for apoptosis induction. Eur J Cancer 2005; 41:1195-202. [PMID: 15911244 DOI: 10.1016/j.ejca.2005.02.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2005] [Accepted: 02/07/2005] [Indexed: 12/11/2022]
Abstract
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and antibodies against TRAIL receptors death receptor 4 (DR4) and death receptor 5 (DR5) are under investigation for cancer therapy. To study the potential application of these agents, the expression of DR4 and DR5 were studied immunohistochemically in colorectal adenomas and carcinomas from patients with sporadic disease (n=74 and 56, respectively), familial adenomatous polyposis (FAP, n=41 and 4, respectively) and hereditary non-polyposis colorectal cancer (HNPCC, n=50 and 21, respectively). BAX, which is frequently mutated in tumours with high-frequency microsatellite instability (MSI-H) may play a role in sensitivity to TRAIL. Therefore, MSI-H carcinomas (n=42, of which 27 sporadic and 15 HNPCC) were analysed for apoptotic activity, assessed by M30 immunoreactivity, and BAX mutations. Most adenomas from all three patient groups expressed DR4 and DR5. Most carcinomas expressed DR4, except for six cases, all with mucinous histology. All carcinomas, including mucinous carcinomas, showed DR5 expression. BAX mutations were found in 6/42 MSI-H cancers with similar apoptotic indices and expression of DR4, DR5 and TRAIL in BAX mutant and wild-type cases. Since most sporadic and hereditary colorectal neoplasms express DR4 and DR5, targeting of these receptors may be a potential prevention or treatment strategy.
Collapse
Affiliation(s)
- Jan J Koornstra
- Department of Gastroenterology, University of Groningen Medical Centre, P.O. Box 30.001, Groningen, RB 9700, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Jalving M, Koornstra JJ, De Jong S, De Vries EGE, Kleibeuker JH. Review article: the potential of combinational regimen with non-steroidal anti-inflammatory drugs in the chemoprevention of colorectal cancer. Aliment Pharmacol Ther 2005; 21:321-39. [PMID: 15709983 DOI: 10.1111/j.1365-2036.2005.02335.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Non-steroidal anti-inflammatory drugs are chemopreventive agents in colorectal cancer. Non-steroidal anti-inflammatory drugs do not, however, offer complete protection against adenoma and carcinoma development. There is increasing interest in combining non-steroidal anti-inflammatory drugs with agents that target specific cell signalling pathways in malignant and premalignant cells. This review aims to describe the current knowledge regarding the efficacy of peroxisome proliferator-activated receptor-gamma ligands, cholesterol synthesis inhibitors (statins), epidermal growth factor signalling inhibitors and tumour necrosis factor-related apoptosis-inducing ligand against colorectal neoplasms and the rationale for combining these drugs with non-steroidal anti-inflammatory drugs to improve efficacy in the chemoprevention of colorectal cancer, a PUBMED computer search of the English language literature was conducted to identify relevant papers published before July 2004. Peroxisome proliferator-activated receptor-gamma ligands and statins, both in clinical use, reduce the growth rate of human colon cancer cells in vitro and in rodents models. In vitro, preclinical in vivo and clinical studies have shown efficacy of epidermal growth factor signalling inhibition in colorectal cancer. In vitro, tumour necrosis factor-related apoptosis-inducing ligand induces apoptosis in human colon cancer cells, but not in normal cells. These drugs have all been shown to interact with non-steroidal anti-inflammatory drugs in colorectal cancer cells and/or in rodent models. Combinational regimen are a promising strategy for the chemoprevention of colorectal cancer and should be further explored.
Collapse
Affiliation(s)
- M Jalving
- Department of Gastroenterology and Hepatology, University Hospital Groningen, Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
45
|
Van Geelen CMM, de Vries EGE, de Jong S. Lessons from TRAIL-resistance mechanisms in colorectal cancer cells: paving the road to patient-tailored therapy. Drug Resist Updat 2005; 7:345-58. [PMID: 15790545 DOI: 10.1016/j.drup.2004.11.002] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2004] [Revised: 11/02/2004] [Accepted: 11/03/2004] [Indexed: 02/06/2023]
Abstract
Colorectal cancer is one of the leading causes of cancer-related deaths worldwide. Intrinsic, as well as acquired, resistance to chemotherapy remains a major problem in the treatment of this disease. It is, therefore, of great importance to develop new, patient-tailored, treatment strategies for colorectal cancer patients. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) acts through the pro-apoptotic DR4 and DR5 receptors in tumor cells without harming normal cells and will soon be tested in clinical trials as a novel anti-cancer agent. However, not all human colon cancer cell lines are sensitive to TRAIL due to intrinsic or acquired TRAIL-resistance. This review discusses the mechanisms and modulation of TRAIL-resistance in colon cancer cells. Cell sensitivity to TRAIL can be affected by TRAIL-receptor expression at the cell membrane, DR4/DR5 ratio and functionality of TRAIL-receptors. Additional intracellular factors leading to TRAIL-resistance affect the caspase 8/c-FLIP ratio, such as loss of caspase 8 and caspase 10 due to mutations or gene methylation, CARP-dependent degradation of active caspase 8 and changes in caspase 8 or c-FLIP expression levels. Further downstream in the TRAIL apoptotic pathway, Bax mutations, or increased expression of IAP family members, in particularly XIAP and survivin, also cause resistance. Chemotherapeutic drugs, NSAIDs, interferon-gamma and proteasome inhibitors can overcome TRAIL-resistance by acting on TRAIL-receptor expression or changing the expression of pro- or anti-apoptotic proteins.
Collapse
Affiliation(s)
- Caroline M M Van Geelen
- Department of Medical Oncology, University Hospital of Groningen, PO Box 30.001, 9700 RB Groningen, Groningen, The Netherlands
| | | | | |
Collapse
|
46
|
Fan QL, Zou WY, Song LH, Wei W. Synergistic antitumor activity of TRAIL combined with chemotherapeutic agents in A549 cell lines in vitro and in vivo. Cancer Chemother Pharmacol 2004; 55:189-96. [PMID: 15290100 DOI: 10.1007/s00280-004-0867-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Accepted: 05/21/2004] [Indexed: 01/03/2023]
Abstract
PURPOSE To investigate the synergistic cytotoxicity of TRAIL in combination with chemotherapeutic agents in A549 cell lines, we systematically evaluated the cytotoxicity of TRAIL alone and TRAIL in combination with cisplatin, paclitaxel (Taxol) or actinomycin D in A549 cell lines in vitro and in vivo, and whether the sensitivity was correlated with the expression level of TRAIL receptors. METHODS We investigated the cytotoxicity of TRAIL alone and the synergistic antitumor effects of TRAIL in combination with chemotherapeutic agents in A549 cells by crystal violet staining and FACS in vitro. The expression levels of DR4, DR5, DcR1 and DcR2 were measured in TRAIL-treated and chemotherapeutic agent-treated A549 cells by Western blotting. The growth inhibition of tumors was evaluated in terms of incidence, volume and weight in a A549-implanted nude mice model. RESULTS Chemotherapeutic agents cisplatin (5.56 mug/ml), Taxol (10 and 30 mug/ml) or actinomycin D (9.26, 83.3 and 750 ng/ml) augmented the cytotoxicity of TRAIL in A549 cell lines within a range of concentrations of TRAIL (1.98-160 ng/ml) in vitro. The expression levels of DR4 and DR5 were not significantly different and the expression of DcR2 was slightly downregulated, but the expression of DcR1 was not detected in non-treated, TRAIL-treated and chemotherapeutic agent-treated A549 cells. The rates of tumor inhibition following treatment with TRAIL alone (15 mg/kg per day, daily for 10 days) and TRAIL/cisplatin (15 mg/kg per day TRAIL, daily for 10 days; 1.5 mg/kg per day cisplatin, daily for 10 days with 7-day intervals) were 28.3% and 76.8% by tumor weight ( P<0.05 for TRAIL alone versus control, P<0.05 for TRAIL/cisplatin versus cisplatin alone and TRAIL alone) on day 65 in vivo. CONCLUSION TRAIL in combination with chemotherapeutic agents cisplatin, Taxol or actinomycin D exerted synergistic antitumor effects in A549 cell lines in vitro and TRAIL/cisplatin demonstrated synergistic antitumor effects in vivo. The expression levels of TRAIL receptors suggested that the synergistic effects of TRAIL in combination with chemotherapeutic agents are not at the receptor level in A549 cell lines.
Collapse
Affiliation(s)
- Qing-Lin Fan
- Institute of Clinical Pharmacology, Anhui Medical University, 230032, Hefei, People's Republic of China
| | | | | | | |
Collapse
|
47
|
Spierings DC, de Vries EG, Vellenga E, van den Heuvel FA, Koornstra JJ, Wesseling J, Hollema H, de Jong S. Tissue distribution of the death ligand TRAIL and its receptors. J Histochem Cytochem 2004; 52:821-31. [PMID: 15150291 DOI: 10.1369/jhc.3a6112.2004] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Recombinant human (rh) TNF-related apoptosis-inducing ligand (TRAIL) harbors potential as an anticancer agent. RhTRAIL induces apoptosis via the TRAIL receptors TRAIL-R1 and TRAIL-R2 in tumors and is non-toxic to nonhuman primates. Because limited data are available about TRAIL receptor distribution, we performed an immunohistochemical (IHC) analysis of the expression of TRAIL-R1, TRAIL-R2, the anti-apoptotic TRAIL receptor TRAIL-R3, and TRAIL in normal human and chimpanzee tissues. In humans, hepatocytes stained positive for TRAIL and TRAIL receptors and bile duct epithelium for TRAIL, TRAIL-R1, and TRAIL-R3. In brains, neurons expressed TRAIL-R1, TRAIL-R2, TRAIL-R3 but no TRAIL. In kidneys, TRAIL-R3 was negative, tubuli contorti expressed TRAIL-R1, TRAIL-R2, and TRAIL, and cells in Henle's loop expressed only TRAIL-R2. Heart myocytes showed positivity for all proteins studied. In colon, TRAIL-R1, TRAIL-R2, and TRAIL were present. Germ and Leydig cells were positive for all proteins studied. Endothelium in liver, heart, kidney, and testis lacked TRAIL-R1 and TRAIL-R2. In alveolar septa and bronchial epithelium TRAIL-R2 was expressed, brain vascular endothelium expressed TRAIL-R2 and TRAIL-R3, and in heart vascular endothelium only TRAIL-R3 was present. Only a few differences were observed between human and chimpanzee liver, brain, and kidney. In contrast to human, chimpanzee bile duct epithelium lacked TRAIL, TRAIL-R1, and TRAIL-R3, lung and colon showed no TRAIL or its receptors, TRAIL-R3 was absent in germ and Leydig cells, and vascular endothelium showed only TRAIL-R2 expression in the brain. In conclusion, comparable expression of TRAIL and TRAIL receptors was observed in human and chimpanzee tissues. Lack of liver toxicity in chimpanzees after rhTRAIL administration despite TRAIL-R1 and TRAIL-R2 expression is reassuring for rhTRAIL application in humans.
Collapse
Affiliation(s)
- Diana C Spierings
- Department of Medical Oncology, University Hospital Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|