1
|
Sun Y, Fang W, Peng J, Liu X, Wang C, Song L, Deng Z. Potential role of CFLAR in enhancing 5-FU sensitivity and modulating immune cell infiltration in breast cancer. Eur J Med Res 2025; 30:265. [PMID: 40211399 PMCID: PMC11983979 DOI: 10.1186/s40001-025-02532-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Breast cancer (BRCA), the most common malignancy among women, is a highly heterogeneous disease. Chemoresistance is a major factor leading to treatment failure in BRCA. However, mechanisms underlying the development of chemoresistance remain unclear. METHODS In this study, we performed a comprehensive bioinformatic analysis to examine the role of cell death-associated genes in BRCA treatment. Specifically, we focused on caspase 8 and Fas-associated protein with death domain-like apoptosis regulator (CFLAR), which was identified as a co-differentially expressed cell death-associated molecule with potential prognostic values. We then validated these findings through in vitro experiments in BT- 549 and MDA-MB- 231 breast cancer cells. RESULTS Based on bioinformatics analysis, CFLAR expression was found to be downregulated in patients with BRCA, whereas its high expression was significantly associated with improved prognosis. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated that aberrantly expressed CFLAR was potentially associated with oxidative phosphorylation, T cell receptor signaling, and NADH dehydrogenase (ubiquinone) activity. In vitro experiments demonstrated that overexpression of CFLAR inhibited the generation of reactive oxygen species (ROS), consequently promoting 5-fluorouracil (5-FU) sensitivity in BT- 549 and MDA-MB- 231 breast cancer cells. The expression of CFLAR was positively correlated with the abundance of several tumor-infiltrating immune cells, especially CD8 + T cells, further supporting the role of CFLAR in immune regulation. CONCLUSION In conclusion, this study reveals the novel roles of CFLAR in enhancing chemotherapy sensitivity and patient outcome in BRCA and underscores its potential as a therapeutic target. These results supported CFLAR as a therapeutic target and prognostic biomarker in BRCA patients.
Collapse
Affiliation(s)
- Yuwei Sun
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Weilun Fang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Xingling Liu
- Department of Pharmacy, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, Guangdong, China.
| | - Chunjiang Wang
- Department of Pharmacy, the Third Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Liying Song
- Department of Pharmacy, the Third Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhenzhen Deng
- Department of Pharmacy, the Third Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| |
Collapse
|
2
|
Lu M, Ren Y, Feng S, Wang S, Xia W, Gu B, Shen Y, Yue A, Li N, Zhang Y, Zhong J. MDM2 inhibitor induces apoptosis in colon cancer cells through activation of the CHOP-DR5 pathway, independent of p53 phenotype. Front Pharmacol 2025; 16:1508421. [PMID: 40264676 PMCID: PMC12011796 DOI: 10.3389/fphar.2025.1508421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction Murine double minute 2 (MDM2), a key negative regulator of p53, forms a feedback loop with p53 to drive tumor progression, including colorectal cancer. Nutlin-3a, an MDM2 inhibitor, induces apoptosis in wild-type p53 tumors, but its effects on p53-mutated cancers and potential p53-independent apoptotic mechanisms remain unclear. Methods We investigated Nutlin-3a's effects on colon cancer cells with varying p53 phenotypes. Endoplasmic reticulum (ER) stress-associated CHOP was detected and knocked down to explore mechanisms. In vitro and in vivo experiments assessed Nutlin-3a's synergy with 5-fluorouracil and TRAIL. Results Nutlin-3a activated caspase-8-dependent extrinsic apoptosis in colon cancer cells via DR5 upregulation, independent of p53 status. ER stress and CHOP activation mediated DR5 induction, driven by calcium release. Combined Nutlin-3a treatment enhanced sensitivity to 5-fluorouracil and TRAIL in vitro and in vivo through caspase-8 pathway activation. Discussion These findings reveal a novel p53-independent apoptotic mechanism of Nutlin-3a involving ER stress and death receptor signaling. This pathway highlights Nutlin-3a's potential as an adjuvant therapy for colon cancer, even in p53-mutated tumors, by enhancing chemotherapeutic efficacy through extrinsic apoptosis.
Collapse
Affiliation(s)
- Manman Lu
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yingli Ren
- College of Medicine, Henan Polytechnic University, Jiaozuo, China
| | - Sijia Feng
- Henan Province Engineering Technology Research Center of Tumor Diagnostic Biomarkers and RNA Interference Drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shenggen Wang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Weiyue Xia
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Baoru Gu
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yuhou Shen
- Department of Abdominal Surgical Oncology Ward 2, Xinxiang Central Hospital, Xinxiang, China
- Department of Abdominal Surgical Oncology Ward 2, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, China
| | - Aimin Yue
- Department of Abdominal Surgical Oncology Ward 2, Xinxiang Central Hospital, Xinxiang, China
- Department of Abdominal Surgical Oncology Ward 2, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, China
| | - Na Li
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yongxi Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Province Engineering Technology Research Center of Tumor Diagnostic Biomarkers and RNA Interference Drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiateng Zhong
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Province Engineering Technology Research Center of Tumor Diagnostic Biomarkers and RNA Interference Drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
3
|
Koolivand Z, Bahreini F, Rayzan E, Rezaei N. Inducing apoptosis in acute myeloid leukemia; mechanisms and limitations. Heliyon 2025; 11:e41355. [PMID: 39811307 PMCID: PMC11730532 DOI: 10.1016/j.heliyon.2024.e41355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Acute myeloid leukemia is the expansion of leukemic stem cells which might originate from a stem cell or a progenitor which has acquired self-renewal capacity. An aggregation of leukemic blasts in bone marrow, peripheral blood, and extramedullary tissue will result in acute myeloid leukemia. The main difficulty in treating acute myeloid leukemia is multidrug resistance, leading to treatment failure. This unfortunate phenomenon is practically elevated because of apoptosis inhibition in tumor cells. Two general apoptotic pathways are the Bcl-2 regulated pathway (the intrinsic pathway) and the death receptor pathway. Deficiencies in each of these apoptotic pathways can cause the usual resistance mechanism in this disease. This article reviews and highlights different antiapoptotic pathways, currently-used treatments, and new findings in this field, which may lead to the development of treatment methods for acute myeloid leukemia.
Collapse
Affiliation(s)
- Zahra Koolivand
- Faculty of Pharmacy and Pharmaceutical Sciences, Islamic Azad University, Tehran Medical Sciences University (IAUTMU), Tehran, Iran
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farbod Bahreini
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Institute of Anatomy, University of Luebeck, Luebeck, Germany
| | - Elham Rayzan
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Guo R, Wei Y, Du Y, Liu L, Zhang H, Ren R, Sun R, Zhang T, Xiong X, Zhao L, Wang H, Guo X, Zhu X. EX527, a sirtuins 1 inhibitor, sensitizes T-cell leukemia to death receptor-mediated apoptosis by downregulating cellular FLICE inhibitory protein. Cancer Biol Ther 2024; 25:2402588. [PMID: 39286953 PMCID: PMC11409494 DOI: 10.1080/15384047.2024.2402588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/22/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Death receptor-mediated extrinsic apoptosis system had been developed as a promising therapeutic strategy in clinical oncology, such as TRAIL therapy. However, multiple studies have demonstrated that TRAIL resistance is the biggest problem for disappointing clinical trials despite preclinical success. Targeting cellular FLICE inhibitory protein (cFLIP) is one strategy of combinatorial therapies to overcome resistance to DR-mediated apoptosis due to its negative regulator of extrinsic apoptosis. E × 527 (Selisistat) is a specific inhibitor of SIRT1 activity with safe and well tolerance in clinical trials. Here, we show that E × 527 could strengthen significantly activation of rhFasL-mediated apoptotic signaling pathway and increased apoptotic rate of T leukemia cells with high expression of cFLIP. Mechanically, Inhibition of SIRT1 by E × 527 increased polyubiquitination level of cFLIP via increasing acetylation of Ku70, which could promote proteosomal degradation of cFLIP protein. It implied that combinatorial therapies of E × 527 plus TRAIL may have a potential as a novel clinical application for TRAIL-resistant hematologic malignancies.
Collapse
Affiliation(s)
- Rongqi Guo
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Yihui Wei
- Henan Red Cross Blood Center, Xinxiang, China
| | - Yating Du
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Luyue Liu
- Departments of Laboratory Medicine, Zhoukou Central Hospital, Zhoukou, China
| | - Haoqi Zhang
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang
Medical University, Xinxiang, China
| | - Ruiying Ren
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Ruili Sun
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Tingting Zhang
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang
Medical University, Xinxiang, China
| | - Xiwen Xiong
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Lijun Zhao
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Hongfei Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Xiaofang Guo
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang
Medical University, Xinxiang, China
| | - Xiaofei Zhu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
5
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
6
|
Gayan S, Teli A, Sonawane A, Dey T. Impact of Chemotherapeutic Stress Depends on The Nature of Breast Cancer Spheroid and Induce Behavioral Plasticity to Resistant Population. Adv Biol (Weinh) 2024; 8:e2300271. [PMID: 38063815 DOI: 10.1002/adbi.202300271] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 11/20/2023] [Indexed: 04/15/2024]
Abstract
Cellular or tumor dormancy, identified recently as one of the main reasons behind post-therapy recurrence, can be caused by diverse reasons. Chemotherapy has recently been recognized as one of such reasons. However, in-depth studies of chemotherapy-induced dormancy are lacking due to the absence of an in vitro human-relevant model tailor-made for such a scenario. This report utilized multicellular breast cancer spheroid to create a primary platform for establishing a chemotherapy-induced dormancy model. It is observed that extreme chemotherapeutic stress affects invasive and non-invasive spheroids differently. Non-invasive spheroids exhibit more resilience and maintain viability and migrational ability, while invasive spheroids display heightened susceptibility and improved tumorigenic capacity. Heterogenous spheroids exhibit increased tumorigenic capacity while show minimal survival ability. Further probing of chemotherapeutically dormant spheroids is needed to understand the molecular mechanism and identify dormancy-related markers to achieve therapeutic success in the future.
Collapse
Affiliation(s)
- Sukanya Gayan
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Abhishek Teli
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Akshay Sonawane
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Tuli Dey
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| |
Collapse
|
7
|
Yaacoub K, Pedeux R, Lafite P, Jarry U, Aci-Sèche S, Bonnet P, Daniellou R, Guillaudeux T. The Identification of New c-FLIP Inhibitors for Restoring Apoptosis in TRAIL-Resistant Cancer Cells. Curr Issues Mol Biol 2024; 46:710-728. [PMID: 38248348 PMCID: PMC10814526 DOI: 10.3390/cimb46010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
The catalytically inactive caspase-8-homologous protein, c-FLIP, is a potent antiapoptotic protein highly expressed in various types of cancers. c-FLIP competes with caspase-8 for binding to the adaptor protein FADD (Fas-Associated Death Domain) following death receptors' (DRs) activation via the ligands of the TNF-R family. As a consequence, the extrinsic apoptotic signaling pathway involving DRs is inhibited. The inhibition of c-FLIP activity in tumor cells might enhance DR-mediated apoptosis and overcome immune and anticancer drug resistance. Based on an in silico approach, the aim of this work was to identify new small inhibitory molecules able to bind selectively to c-FLIP and block its anti-apoptotic activity. Using a homology 3D model of c-FLIP, an in silico screening of 1880 compounds from the NCI database (National Cancer Institute) was performed. Nine molecules were selected for in vitro assays, based on their binding affinity to c-FLIP and their high selectivity compared to caspase-8. These molecules selectively bind to the Death Effector Domain 2 (DED2) of c-FLIP. We have tested in vitro the inhibitory effect of these nine molecules using the human lung cancer cell line H1703, overexpressing c-FLIP. Our results showed that six of these newly identified compounds efficiently prevent FADD/c-FLIP interactions in a molecular pull-down assay, as well as in a DISC immunoprecipitation assay. The overexpression of c-FLIP in H1703 prevents TRAIL-mediated apoptosis; however, a combination of TRAIL with these selected molecules significantly restored TRAIL-induced cell death by rescuing caspase cleavage and activation. Altogether, our findings indicate that new inhibitory chemical molecules efficiently prevent c-FLIP recruitment into the DISC complex, thus restoring the caspase-8-dependent apoptotic cascade. These results pave the way to design new c-FLIP inhibitory molecules that may serve as anticancer agents in tumors overexpressing c-FLIP.
Collapse
Affiliation(s)
- Katherine Yaacoub
- CNRS, INSERM, BIOSIT UAR 3480, US-S018, Rennes University, F-35000 Rennes, France; (K.Y.); (U.J.)
- INSERM, OSS (Oncogenesis Stress Signaling), UMR-S1242, CLCC Eugène Marquis, Rennes University, F-35000 Rennes, France;
| | - Rémy Pedeux
- INSERM, OSS (Oncogenesis Stress Signaling), UMR-S1242, CLCC Eugène Marquis, Rennes University, F-35000 Rennes, France;
| | - Pierre Lafite
- CNRS, ICOA, UMR 7311, Orléans University, F-45067 Orléans, France; (P.L.); (S.A.-S.); (P.B.); (R.D.)
| | - Ulrich Jarry
- CNRS, INSERM, BIOSIT UAR 3480, US-S018, Rennes University, F-35000 Rennes, France; (K.Y.); (U.J.)
| | - Samia Aci-Sèche
- CNRS, ICOA, UMR 7311, Orléans University, F-45067 Orléans, France; (P.L.); (S.A.-S.); (P.B.); (R.D.)
| | - Pascal Bonnet
- CNRS, ICOA, UMR 7311, Orléans University, F-45067 Orléans, France; (P.L.); (S.A.-S.); (P.B.); (R.D.)
| | - Richard Daniellou
- CNRS, ICOA, UMR 7311, Orléans University, F-45067 Orléans, France; (P.L.); (S.A.-S.); (P.B.); (R.D.)
| | - Thierry Guillaudeux
- CNRS, INSERM, BIOSIT UAR 3480, US-S018, Rennes University, F-35000 Rennes, France; (K.Y.); (U.J.)
- INSERM, OSS (Oncogenesis Stress Signaling), UMR-S1242, CLCC Eugène Marquis, Rennes University, F-35000 Rennes, France;
| |
Collapse
|
8
|
Kumar K, Rani V, Mishra M, Chawla R. New paradigm in combination therapy of siRNA with chemotherapeutic drugs for effective cancer therapy. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100103. [PMID: 35586474 PMCID: PMC9108887 DOI: 10.1016/j.crphar.2022.100103] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 12/11/2022] Open
Abstract
Chemotherapeutics drugs play a pivotal role in the treatment of cancer. However, many issues generate by chemotherapy drugs, including unfavorable harm to healthy cells and multidrug resistance (MDR), persist and have a negative impact on therapeutic outcomes. When compared to monotherapy, combination cancer therapy has many advantages, like improving efficacy through synergistic effects and overcoming drug resistance. Combination treatment may comprise several chemotherapeutics drugs and combinations of chemotherapeutic drugs with some other therapeutic options such as surgery or radiation. Cancer treatment that utilizes co-delivery strategies with siRNA and chemotherapeutic drugs has been shown to have highly effective antitumor effects in the treatment of many cancers. However, the highly complex mechanisms of chemotherapeutic drugs-siRNA pairs during the co-delivery process have received little attention. The ideal combination of chemotherapeutic drugs with siRNA is very crucial for producing the desirable anticancer effects that would greatly enhance therapeutic efficiency. This review puts an emphasis on the logic for choosing suitable chemotherapeutic drug-siRNA combinations, which may open the way for the co-delivery of chemotherapeutic drugs and siRNA for treating cancer in the clinic. This review summarizes recent breakthrough in the area of diverse mechanism-based chemotherapeutic drugs-siRNA combinations in cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Ruchi Chawla
- Corresponding author. Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, U.P., India.
| |
Collapse
|
9
|
Nanomedicine in Pancreatic Cancer: Current Status and Future Opportunities for Overcoming Therapy Resistance. Cancers (Basel) 2021; 13:cancers13246175. [PMID: 34944794 PMCID: PMC8699181 DOI: 10.3390/cancers13246175] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Despite access to a vast arsenal of anticancer agents, many fail to realise their full therapeutic potential in clinical practice. One key determinant of this is the evolution of multifaceted resistance mechanisms within the tumour that may either pre-exist or develop during the course of therapy. This is particularly evident in pancreatic cancer, where limited responses to treatment underlie dismal survival rates, highlighting the urgent need for new therapeutic approaches. Here, we discuss the major features of pancreatic tumours that contribute to therapy resistance, and how they may be alleviated through exploitation of the mounting and exciting promise of nanomedicines; a unique collection of nanoscale platforms with tunable and multifunctional capabilities that have already elicited a widespread impact on cancer management. Abstract The development of drug resistance remains one of the greatest clinical oncology challenges that can radically dampen the prospect of achieving complete and durable tumour control. Efforts to mitigate drug resistance are therefore of utmost importance, and nanotechnology is rapidly emerging for its potential to overcome such issues. Studies have showcased the ability of nanomedicines to bypass drug efflux pumps, counteract immune suppression, serve as radioenhancers, correct metabolic disturbances and elicit numerous other effects that collectively alleviate various mechanisms of tumour resistance. Much of this progress can be attributed to the remarkable benefits that nanoparticles offer as drug delivery vehicles, such as improvements in pharmacokinetics, protection against degradation and spatiotemporally controlled release kinetics. These attributes provide scope for precision targeting of drugs to tumours that can enhance sensitivity to treatment and have formed the basis for the successful clinical translation of multiple nanoformulations to date. In this review, we focus on the longstanding reputation of pancreatic cancer as one of the most difficult-to-treat malignancies where resistance plays a dominant role in therapy failure. We outline the mechanisms that contribute to the treatment-refractory nature of these tumours, and how they may be effectively addressed by harnessing the unique capabilities of nanomedicines. Moreover, we include a brief perspective on the likely future direction of nanotechnology in pancreatic cancer, discussing how efforts to develop multidrug formulations will guide the field further towards a therapeutic solution for these highly intractable tumours.
Collapse
|
10
|
Hamilton C, Fox JP, Longley DB, Higgins CA. Therapeutics Targeting the Core Apoptotic Machinery. Cancers (Basel) 2021; 13:cancers13112618. [PMID: 34073507 PMCID: PMC8198123 DOI: 10.3390/cancers13112618] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/09/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Cancer develops when the balance between cell death and cell division in tissues is dysregulated. A key focus of cancer drug discovery is identifying therapeutic agents which will selectively kill and eliminate cancer cells from the body. A number of proteins can prevent the death of cancer cells and developing inhibitors against these proteins to promote cancer cell death is a focus of recent drug discovery efforts. This review aims to summarize the key targets being explored, the drug development approaches being adopted, and the success or limitations of agents currently approved or in clinical development. Abstract Therapeutic targeting of the apoptotic pathways for the treatment of cancer is emerging as a valid and exciting approach in anti-cancer therapeutics. Accumulating evidence demonstrates that cancer cells are typically “addicted” to a small number of anti-apoptotic proteins for their survival, and direct targeting of these proteins could provide valuable approaches for directly killing cancer cells. Several approaches and agents are in clinical development targeting either the intrinsic mitochondrial apoptotic pathway or the extrinsic death receptor mediated pathways. In this review, we discuss the main apoptosis pathways and the key molecular targets which are the subject of several drug development approaches, the clinical development of these agents and the emerging resistance factors and combinatorial treatment approaches for this class of agents with existing and emerging novel targeted anti-cancer therapeutics.
Collapse
|
11
|
Supper E, Rudat S, Iyer V, Droop A, Wong K, Spinella JF, Thomas P, Sauvageau G, Adams DJ, Wong CC. Cut-like homeobox 1 (CUX1) tumor suppressor gene haploinsufficiency induces apoptosis evasion to sustain myeloid leukemia. Nat Commun 2021; 12:2482. [PMID: 33931647 PMCID: PMC8087769 DOI: 10.1038/s41467-021-22750-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/24/2021] [Indexed: 01/19/2023] Open
Abstract
While oncogenes promote tumorigenesis, they also induce deleterious cellular stresses, such as apoptosis, that cancer cells must combat by coopting adaptive responses. Whether tumor suppressor gene haploinsufficiency leads to such phenomena and their mechanistic basis is unclear. Here, we demonstrate that elevated levels of the anti-apoptotic factor, CASP8 and FADD-like apoptosis regulator (CFLAR), promotes apoptosis evasion in acute myeloid leukemia (AML) cells haploinsufficient for the cut-like homeobox 1 (CUX1) transcription factor, whose loss is associated with dismal clinical prognosis. Genome-wide CRISPR/Cas9 screening identifies CFLAR as a selective, acquired vulnerability in CUX1-deficient AML, which can be mimicked therapeutically using inhibitor of apoptosis (IAP) antagonists in murine and human AML cells. Mechanistically, CUX1 deficiency directly alleviates CUX1 repression of the CFLAR promoter to drive CFLAR expression and leukemia survival. These data establish how haploinsufficiency of a tumor suppressor is sufficient to induce advantageous anti-apoptosis cell survival pathways and concurrently nominate CFLAR as potential therapeutic target in these poor-prognosis leukemias.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- CASP8 and FADD-Like Apoptosis Regulating Protein/genetics
- CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism
- Cell Cycle/drug effects
- Cell Cycle/genetics
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Cell Survival/genetics
- Chromatin Immunoprecipitation
- Dipeptides/pharmacology
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Gene Ontology
- Genes, Tumor Suppressor
- Haploinsufficiency
- Hematopoietic Stem Cells/metabolism
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Indoles/pharmacology
- Kaplan-Meier Estimate
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myelomonocytic, Chronic/genetics
- Leukemia, Myelomonocytic, Chronic/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mutation
- Nuclear Proteins/deficiency
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Promoter Regions, Genetic
- Protein Array Analysis
- Repressor Proteins/deficiency
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
Collapse
Affiliation(s)
- Emmanuelle Supper
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Saskia Rudat
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Vivek Iyer
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Alastair Droop
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Kim Wong
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Jean-François Spinella
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, 2950 Chemin de Polytechnique Pavillon, Marcelle-Coutu, Montréal, QC, Canada
| | - Patrick Thomas
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Guy Sauvageau
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, 2950 Chemin de Polytechnique Pavillon, Marcelle-Coutu, Montréal, QC, Canada
| | - David J Adams
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Chi C Wong
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, UK.
- Department of Haematology, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
12
|
Tyagi A, Chandrasekaran B, Kolluru V, Baby BV, Sripathi CA, Ankem MK, Ramisetti SR, Chirasani VR, Dokholyan NV, Sharma AK, Damodaran C. ASR490, a Small Molecule, Overrides Aberrant Expression of Notch1 in Colorectal Cancer. Mol Cancer Ther 2020; 19:2422-2431. [PMID: 33087513 PMCID: PMC10694926 DOI: 10.1158/1535-7163.mct-19-0949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 05/14/2020] [Accepted: 10/05/2020] [Indexed: 11/16/2022]
Abstract
Notch1 activation triggers significant oncogenic signaling that manifests as enhanced metastatic potential and tumorigenesis in colorectal cancer. Novel small-molecule inhibitors, mainly plant-derived analogs, have low toxicity profiles and higher bioavailability. In this study, we have developed a small molecule, ASR490, by modifying structure of naturally occurring compound Withaferin A. ASR490 showed a growth-inhibitory potential by downregulating Notch1 signaling in HCT116 and SW620 cell lines. Docking studies and thermal shift assays confirmed that ASR490 binds to Notch1, whereas no changes in Notch2 and Notch3 expression were seen in colorectal cancer cells. Notch1 governs epithelial-to-mesenchymal transition signaling and is responsible for metastasis, which was abolished by ASR490 treatment. To further confirm the therapeutic potential of ASR490, we stably overexpressed Notch1 in HCT-116 cells and determined its inhibitory potential in transfected colorectal cancer (Notch1/HCT116) cells. ASR490 effectively prevented cell growth in both the vector (P = 0.005) and Notch1 (P = 0.05) transfectants. The downregulation of Notch1 signaling was evident, which corresponded with downregulation of mesenchymal markers, including N-cadherin and β-catenin and induction of E-cadherin in HCT-116 transfectants. Intraperitoneal administration of a 1% MTD dose of ASR490 (5 mg/kg) effectively suppressed the tumor growth in control (pCMV/HCT116) and Notch1/HCT116 in xenotransplanted mice. In addition, downregulation of Notch1 and survival signaling in ASR-treated tumors confirmed the in vitro results. In conclusion, ASR490 appears to be a potent agent that can inhibit Notch1 signaling in colorectal cancer.
Collapse
Affiliation(s)
- Ashish Tyagi
- Department of Urology, University of Louisville, Louisville, Kentucky
| | | | - Venkatesh Kolluru
- Department of Urology, University of Louisville, Louisville, Kentucky
| | - Becca V Baby
- Department of Urology, University of Louisville, Louisville, Kentucky
| | - Cibi A Sripathi
- Department of Urology, University of Louisville, Louisville, Kentucky
| | - Murali K Ankem
- Department of Urology, University of Louisville, Louisville, Kentucky
| | - Srinivasa R Ramisetti
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, Pennsylvania
| | - Venkat R Chirasani
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, Pennsylvania
| | - Nikolay V Dokholyan
- Department of Pharmacology, Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, Pennsylvania
| | - Chendil Damodaran
- Department of Urology, University of Louisville, Louisville, Kentucky.
| |
Collapse
|
13
|
Lees A, McIntyre AJ, Crawford NT, Falcone F, McCann C, Holohan C, Quinn GP, Roberts JZ, Sessler T, Gallagher PF, Gregg GMA, McAllister K, McLaughlin KM, Allen WL, Egan LJ, Ryan AE, Labonte-Wilson MJ, Dunne PD, Wappett M, Coyle VM, Johnston PG, Kerr EM, Longley DB, McDade SS. The pseudo-caspase FLIP(L) regulates cell fate following p53 activation. Proc Natl Acad Sci U S A 2020; 117:17808-17819. [PMID: 32661168 PMCID: PMC7395556 DOI: 10.1073/pnas.2001520117] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
p53 is the most frequently mutated, well-studied tumor-suppressor gene, yet the molecular basis of the switch from p53-induced cell-cycle arrest to apoptosis remains poorly understood. Using a combination of transcriptomics and functional genomics, we unexpectedly identified a nodal role for the caspase-8 paralog and only human pseudo-caspase, FLIP(L), in regulating this switch. Moreover, we identify FLIP(L) as a direct p53 transcriptional target gene that is rapidly up-regulated in response to Nutlin-3A, an MDM2 inhibitor that potently activates p53. Genetically or pharmacologically inhibiting expression of FLIP(L) using siRNA or entinostat (a clinically relevant class-I HDAC inhibitor) efficiently promoted apoptosis in colorectal cancer cells in response to Nutlin-3A, which otherwise predominantly induced cell-cycle arrest. Enhanced apoptosis was also observed when entinostat was combined with clinically relevant, p53-activating chemotherapy in vitro, and this translated into enhanced in vivo efficacy. Mechanistically, FLIP(L) inhibited p53-induced apoptosis by blocking activation of caspase-8 by the TRAIL-R2/DR5 death receptor; notably, this activation was not dependent on receptor engagement by its ligand, TRAIL. In the absence of caspase-8, another of its paralogs, caspase-10 (also transcriptionally up-regulated by p53), induced apoptosis in Nutlin-3A-treated, FLIP(L)-depleted cells, albeit to a lesser extent than in caspase-8-proficient cells. FLIP(L) depletion also modulated transcription of canonical p53 target genes, suppressing p53-induced expression of the cell-cycle regulator p21 and enhancing p53-induced up-regulation of proapoptotic PUMA. Thus, even in the absence of caspase-8/10, FLIP(L) silencing promoted p53-induced apoptosis by enhancing PUMA expression. Thus, we report unexpected, therapeutically relevant roles for FLIP(L) in determining cell fate following p53 activation.
Collapse
Affiliation(s)
- Andrea Lees
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Alexander J McIntyre
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Nyree T Crawford
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Fiammetta Falcone
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Christopher McCann
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Caitriona Holohan
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Gerard P Quinn
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Jamie Z Roberts
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Tamas Sessler
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Peter F Gallagher
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Gemma M A Gregg
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Katherine McAllister
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Kirsty M McLaughlin
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Wendy L Allen
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Laurence J Egan
- Discipline of Pharmacology & Therapeutics, Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Aideen E Ryan
- Discipline of Pharmacology & Therapeutics, Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
- Regenerative Medicine Institute, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Melissa J Labonte-Wilson
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Philip D Dunne
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Mark Wappett
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Vicky M Coyle
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Patrick G Johnston
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Emma M Kerr
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom
| | - Daniel B Longley
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom;
| | - Simon S McDade
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland BT9 7BL, United Kingdom;
| |
Collapse
|
14
|
Ferris J, Espona-Fiedler M, Hamilton C, Holohan C, Crawford N, McIntyre AJ, Roberts JZ, Wappett M, McDade SS, Longley DB, Coyle V. Pevonedistat (MLN4924): mechanism of cell death induction and therapeutic potential in colorectal cancer. Cell Death Discov 2020; 6:61. [PMID: 32714568 PMCID: PMC7374701 DOI: 10.1038/s41420-020-00296-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/15/2020] [Accepted: 06/30/2020] [Indexed: 12/25/2022] Open
Abstract
Pevonedistat (MLN4924), a selective inhibitor of the NEDD8-activating enzyme E1 regulatory subunit (NAE1), has demonstrated significant therapeutic potential in several malignancies. Although multiple mechanisms-of-action have been identified, how MLN4924 induces cell death and its potential as a combinatorial agent with standard-of-care (SoC) chemotherapy in colorectal cancer (CRC) remains largely undefined. In an effort to understand MLN4924-induced cell death in CRC, we identified p53 as an important mediator of the apoptotic response to MLN4924. We also identified roles for the extrinsic (TRAIL-R2/caspase-8) and intrinsic (BAX/BAK) apoptotic pathways in mediating the apoptotic effects of MLN4924 in CRC cells, as well as a role for BID, which modulates a cross-talk between these pathways. Depletion of the anti-apoptotic protein FLIP, which we identify as a novel mediator of resistance to MLN4924, enhanced apoptosis in a p53-, TRAIL-R2/DR5-, and caspase-8-dependent manner. Notably, TRAIL-R2 was involved in potentiating the apoptotic response to MLN4924 in the absence of FLIP, in a ligand-independent manner. Moreoever, when paired with SoC chemotherapies, MLN4924 demonstrated synergy with the irinotecan metabolite SN38. The cell death induced by MLN4924/SN38 combination was dependent on activation of mitochondria through BAX/BAK, but in a p53-independent manner, an important observation given the high frequency of TP53 mutation(s) in advanced CRC. These results uncover mechanisms of cell death induced by MLN4924 and suggest that this second-generation proteostasis-disrupting agent may have its most widespread activity in CRC, in combination with irinotecan-containing treatment regimens.
Collapse
Affiliation(s)
- Jennifer Ferris
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland BT9 7BL UK
| | - Margarita Espona-Fiedler
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland BT9 7BL UK
| | - Claudia Hamilton
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland BT9 7BL UK
| | - Caitriona Holohan
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland BT9 7BL UK
| | - Nyree Crawford
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland BT9 7BL UK
| | - Alex J. McIntyre
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland BT9 7BL UK
| | - Jamie Z. Roberts
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland BT9 7BL UK
| | - Mark Wappett
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland BT9 7BL UK
| | - Simon S. McDade
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland BT9 7BL UK
| | - Daniel B. Longley
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland BT9 7BL UK
| | - Victoria Coyle
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland BT9 7BL UK
| |
Collapse
|
15
|
Sordo-Bahamonde C, Lorenzo-Herrero S, Payer ÁR, Gonzalez S, López-Soto A. Mechanisms of Apoptosis Resistance to NK Cell-Mediated Cytotoxicity in Cancer. Int J Mol Sci 2020; 21:ijms21103726. [PMID: 32466293 PMCID: PMC7279491 DOI: 10.3390/ijms21103726] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cells are major contributors to immunosurveillance and control of tumor development by inducing apoptosis of malignant cells. Among the main mechanisms involved in NK cell-mediated cytotoxicity, the death receptor pathway and the release of granules containing perforin/granzymes stand out due to their efficacy in eliminating tumor cells. However, accumulated evidence suggest a profound immune suppression in the context of tumor progression affecting effector cells, such as NK cells, leading to decreased cytotoxicity. This diminished capability, together with the development of resistance to apoptosis by cancer cells, favor the loss of immunogenicity and promote immunosuppression, thus partially inducing NK cell-mediated killing resistance. Altered expression patterns of pro- and anti-apoptotic proteins along with genetic background comprise the main mechanisms of resistance to NK cell-related apoptosis. Herein, we summarize the main effector cytotoxic mechanisms against tumor cells, as well as the major resistance strategies acquired by tumor cells that hamper the extrinsic and intrinsic apoptotic pathways related to NK cell-mediated killing.
Collapse
Affiliation(s)
- Christian Sordo-Bahamonde
- Department of Functional Biology, Immunology, University of Oviedo, 33006 Oviedo, Spain; (S.L.-H.); (S.G.)
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Correspondence: (C.S.-B.); (A.L.-S.)
| | - Seila Lorenzo-Herrero
- Department of Functional Biology, Immunology, University of Oviedo, 33006 Oviedo, Spain; (S.L.-H.); (S.G.)
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Ángel R. Payer
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Hematology, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
| | - Segundo Gonzalez
- Department of Functional Biology, Immunology, University of Oviedo, 33006 Oviedo, Spain; (S.L.-H.); (S.G.)
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Alejandro López-Soto
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Biochemistry and Molecular Biology, University of Oviedo, 33006 Oviedo, Spain
- Correspondence: (C.S.-B.); (A.L.-S.)
| |
Collapse
|
16
|
Su CM, Weng YS, Kuan LY, Chen JH, Hsu FT. Suppression of PKCδ/NF-κB Signaling and Apoptosis Induction through Extrinsic/Intrinsic Pathways Are Associated Magnolol-Inhibited Tumor Progression in Colorectal Cancer In Vitro and In Vivo. Int J Mol Sci 2020; 21:ijms21103527. [PMID: 32429376 PMCID: PMC7278962 DOI: 10.3390/ijms21103527] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
Magnolol is one of the hydroxylated biphenyl compounds from the root and stem bark of Magnolia officinalis, which shown to possess anti-colorectal cancer (CRC) effects. However, the regulatory mechanism of magnolol on apoptosis and NF-κB signaling in human CRC has not been elucidated. Thus, we investigated the inhibitory mechanism of magnolol on human and mouse CRC (HT-29 and CT-26) in vitro and in vivo. Results from reporter gene assay indicated that both magnolol and rottlerin (PKCδ inhibitor) reduced the endogenous NF-κB activity. In addition, indolactam V (PKCδ activator)-induced NF-κB signaling was significantly suppressed with both magnolol and rottlerin treatment. Results from Western blotting also indicated that phosphorylation of PKCδ and NF-κB -related proteins involved in tumor progression were effectively decreased by magnolol treatment. The invasion capacity of CRC cells was also attenuated by both magnolol and rottlerin. Furthermore, magnolol triggered Fas/Fas-L mediated extrinsic apoptosis and mitochondria mediated intrinsic apoptosis were validated by flow cytometry. Most importantly, tumor growth in both HT-29 and CT-26 bearing mice were suppressed by magnolol, but no pathologic change was detected in mice kidney, spleen, and liver. As confirmed by immunohistochemistry (IHC) staining from tumor tissue, PKCδ/NF-κB signaling and downstream proteins expression were decreased, while apoptotic proteins expression was increased in the magnolol treated group. According to these results, we suggest that the induction of apoptosis through extrinsic/intrinsic pathways and the blockage of PKCδ/NF-κB signaling are associated with the magnolol-inhibited progression of CRC.
Collapse
Affiliation(s)
- Chun-Min Su
- Department of Surgery, Show Chwan Memorial Hospital, Changhua 500, Taiwan;
| | - Yueh-Shan Weng
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan;
| | - Lin-Yen Kuan
- Department of Emergency Medicine, Cathay General Hospital, Taipei 106, Taiwan; (L.-Y.K.); (J.-H.C.)
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Jiann-Hwa Chen
- Department of Emergency Medicine, Cathay General Hospital, Taipei 106, Taiwan; (L.-Y.K.); (J.-H.C.)
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan;
- Correspondence: or ; Tel.: +886-4-22053366 (ext. 2532)
| |
Collapse
|
17
|
Smyth P, Sessler T, Scott CJ, Longley DB. FLIP(L): the pseudo-caspase. FEBS J 2020; 287:4246-4260. [PMID: 32096279 PMCID: PMC7586951 DOI: 10.1111/febs.15260] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/10/2020] [Accepted: 02/24/2020] [Indexed: 12/27/2022]
Abstract
Possessing structural homology with their active enzyme counterparts but lacking catalytic activity, pseudoenzymes have been identified for all major enzyme groups. Caspases are a family of cysteine‐dependent aspartate‐directed proteases that play essential roles in regulating cell death and inflammation. Here, we discuss the only human pseudo‐caspase, FLIP(L), a paralog of the apoptosis‐initiating caspases, caspase‐8 and caspase‐10. FLIP(L) has been shown to play a key role in regulating the processing and activity of caspase‐8, thereby modulating apoptotic signaling mediated by death receptors (such as TRAIL‐R1/R2), TNF receptor‐1 (TNFR1), and Toll‐like receptors. In this review, these canonical roles of FLIP(L) are discussed. Additionally, a range of nonclassical pseudoenzyme roles are described, in which FLIP(L) functions independently of caspase‐8. These nonclassical pseudoenzyme functions enable FLIP(L) to play key roles in the regulation of a wide range of biological processes beyond its canonical roles as a modulator of cell death.
Collapse
Affiliation(s)
- Peter Smyth
- The Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| | - Tamas Sessler
- The Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| | - Christopher J Scott
- The Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| | - Daniel B Longley
- The Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| |
Collapse
|
18
|
Humphreys LM, Fox JP, Higgins CA, Majkut J, Sessler T, McLaughlin K, McCann C, Roberts JZ, Crawford NT, McDade SS, Scott CJ, Harrison T, Longley DB. A revised model of TRAIL-R2 DISC assembly explains how FLIP(L) can inhibit or promote apoptosis. EMBO Rep 2020; 21:e49254. [PMID: 32009295 PMCID: PMC7054686 DOI: 10.15252/embr.201949254] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/20/2019] [Accepted: 01/13/2020] [Indexed: 11/23/2022] Open
Abstract
The long FLIP splice form FLIP(L) can act as both an inhibitor and promoter of caspase‐8 at death‐inducing signalling complexes (DISCs) formed by death receptors such as TRAIL‐R2 and related intracellular complexes such as the ripoptosome. Herein, we describe a revised DISC assembly model that explains how FLIP(L) can have these opposite effects by defining the stoichiometry (with respect to caspase‐8) at which it converts from being anti‐ to pro‐apoptotic at the DISC. We also show that in the complete absence of FLIP(L), procaspase‐8 activation at the TRAIL‐R2 DISC has significantly slower kinetics, although ultimately the extent of apoptosis is significantly greater. This revised model of DISC assembly also explains why FLIP's recruitment to the TRAIL‐R2 DISC is impaired in the absence of caspase‐8 despite showing that it can interact with the DISC adaptor protein FADD and why the short FLIP splice form FLIP(S) is the more potent inhibitor of DISC‐mediated apoptosis.
Collapse
Affiliation(s)
- Luke M Humphreys
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Jennifer P Fox
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Catherine A Higgins
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Joanna Majkut
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Tamas Sessler
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Kirsty McLaughlin
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Christopher McCann
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Jamie Z Roberts
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Nyree T Crawford
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Simon S McDade
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Christopher J Scott
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Timothy Harrison
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Daniel B Longley
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| |
Collapse
|
19
|
Kiany S, Harrison D, Gordon N. The Histone Deacetylase Inhibitor Entinostat/Syndax 275 in Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1257:75-83. [PMID: 32483732 DOI: 10.1007/978-3-030-43032-0_7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The prognosis for metastatic osteosarcoma (OS) is poor and has not changed in several decades. Therapeutic paradigms that target and exploit novel molecular pathways are desperately needed. Recent preclinical data suggests that modulation of the Fas/FasL pathway may offer benefit in the treatment of refractory osteosarcoma. Fas and FasL are complimentary receptor-ligand proteins. Fas is expressed in multiple tissues, whereas FasL is restricted to privilege organs, such as the lung. Fas expression has been shown to inversely correlate with the metastatic potential of OS cells; tumor cells which express high levels of Fas have decreased metastatic potential and the ones that reach the lung undergo cell death upon interaction with constitutive FasL in the lung. Agents such as gemcitabine and the HDAC inhibitor, entinostat/Syndax 275, have been shown to upregulate Fas expression on OS cells, potentially leading to decreased OS pulmonary metastasis and improved outcome. Clinical trials are in development to evaluate this combination as a potential treatment option for patients with refractory OS.
Collapse
Affiliation(s)
- Simin Kiany
- Department of Pediatrics Research, MD Anderson Cancer Center, Houston, TX, USA
| | - Douglas Harrison
- Department of Pediatrics - Patient Care, MD Anderson Cancer Center, Houston, TX, USA
| | - Nancy Gordon
- Department of Pediatrics Research, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
20
|
Zong H, Zhou H, Xiang Y, Wu G. miR-125b suppresses cellular proliferation by targeting c-FLIP in gallbladder carcinoma. Oncol Lett 2019; 18:6822-6828. [PMID: 31788125 DOI: 10.3892/ol.2019.11000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/13/2019] [Indexed: 01/07/2023] Open
Abstract
Gallbladder carcinoma (GBC) is the most common malignant tumor of the biliary tract. The incidence rate of gallbladder cancer ranks sixth among gastrointestinal types of cancer, and its incidence is increasing each year. Further clarification of the pathogenesis of GBC is essential, and identification of novel effective treatments is required. It has been previously demonstrated that high expression of the anti-apoptotic protein cellular Fas-associated death domain-like interleukin-1-converting enzyme inhibitory protein (c-FLIP) in GBC inhibited apoptosis in gallbladder cancer cells. In subsequent experiments, it was observed that microRNA (miR)-125b could target c-FLIP and inhibit the protein expression of c-FLIP by binding to the 3'untranslated regions of c-FLIP mRNA. In addition, the expression of miR-125b in GBC was significantly decreased, and the growth of gallbladder cancer cells was inhibited by the overexpression of miR-125b. The present study demonstrated that miR-125b could suppress the proliferation of gallbladder cancer cells by targeting c-FLIP. c-FLIP enriched the target gene pathway of miR-125b and may serve as a novel target for the treatment of GBC.
Collapse
Affiliation(s)
- Huajie Zong
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Huading Zhou
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Yang Xiang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Gang Wu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
21
|
Negative regulators of cell death pathways in cancer: perspective on biomarkers and targeted therapies. Apoptosis 2019; 23:93-112. [PMID: 29322476 DOI: 10.1007/s10495-018-1440-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cancer is a primary cause of human fatality and conventional cancer therapies, e.g., chemotherapy, are often associated with adverse side-effects, tumor drug-resistance, and recurrence. Molecularly targeted therapy, composed of small-molecule inhibitors and immunotherapy (e.g., monoclonal antibody and cancer vaccines), is a less harmful alternative being more effective against cancer cells whilst preserving healthy tissues. Drug-resistance, however, caused by negative regulation of cell death signaling pathways, is still a challenge. Circumvention of negative regulators of cell death pathways or development of predictive and response biomarkers is, therefore, quintessential. This review critically discusses the current state of knowledge on targeting negative regulators of cell death signaling pathways including apoptosis, ferroptosis, necroptosis, autophagy, and anoikis and evaluates the recent advances in clinical and preclinical research on biomarkers of negative regulators. It aims to provide a comprehensive platform for designing efficacious polytherapies including novel agents for restoring cell death signaling pathways or targeting alternative resistance pathways to improve the chances for antitumor responses. Overall, it is concluded that nonapoptotic cell death pathways are a potential research arena for drug discovery, development of novel biomarkers and targeted therapies.
Collapse
|
22
|
Rong G, Yang X, Wu H, Wu Y. miR-150-504-519d inhibits the growth of human colorectal cancer cell line SW48 and downregulates c-FLIP receptor. J Cell Biochem 2019; 120:7962-7969. [PMID: 30548660 DOI: 10.1002/jcb.28073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/22/2018] [Indexed: 01/24/2023]
Abstract
microRNAs (miRNAs) are noncoding RNAs that regulates the expression of target messenger RNAs (mRNAs). c-FLIP is an inhibitor of cell apoptosis through inhibition of caspase 8. miR-150, miR-504, and miR-519d were related to cancer cell proliferation, invasion, and migration in colorectal cancer (CRC). However, the role of miR-150-504-519d in CRC has not been studied and the relationship between miR-150-504-519d and c-FLIP remains unclear. In this study, we found that c-FLIP was upregulated in CRC tissues, without detectable expression in normal CRC tissues. Using SW48 cell line, we further showed that miR-150-504-519d inhibited migration, invasion, and promoted apoptosis of SW48 cells. Moreover, in SW48 cell line transfected with miR-150-504-519d, the protein expression of c-FLIP was significantly lower compared with cells transfected with scramble. Our results demonstrated upregulation of c-FLIP in CRC, which was downregulated in SW48 cells after the transfection of miR-150-504-519d, suggesting that manipulation of miR-150-504-519d expression might be a novel approach for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Guoqiang Rong
- Department of General Surgery, The Fifth People s Hospital of Changshu, Changshu, Jiangsu, P.R. China
| | - Xiaodong Yang
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Haorong Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Yongyou Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
23
|
Safa AR, Kamocki K, Saadatzadeh MR, Bijangi-Vishehsaraei K. c-FLIP, a Novel Biomarker for Cancer Prognosis, Immunosuppression, Alzheimer's Disease, Chronic Obstructive Pulmonary Disease (COPD), and a Rationale Therapeutic Target. BIOMARKERS JOURNAL 2019; 5:4. [PMID: 32352084 PMCID: PMC7189798 DOI: 10.36648/2472-1646.5.1.59] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dysregulation of c-FLIP (cellular FADD-like IL-1β-converting enzyme inhibitory protein) has been shown in several diseases including cancer, Alzheimer's disease, and chronic obstructive pulmonary disease (COPD). c-FLIP is a critical anti-cell death protein often overexpressed in tumors and hematological malignancies and its increased expression is often associated with a poor prognosis. c-FLIP frequently exists as long (c-FLIPL) and short (c-FLIPS) isoforms, regulates its anti-cell death functions through binding to FADD (FAS associated death domain protein), an adaptor protein known to activate caspases-8 and -10 and links c-FLIP to several cell death regulating complexes including the death-inducing signaling complex (DISC) formed by various death receptors. c-FLIP also plays a critical role in necroptosis and autophagy. Furthermore, c-FLIP is able to activate several pathways involved in cytoprotection, proliferation, and survival of cancer cells through various critical signaling proteins. Additionally, c-FLIP can inhibit cell death induced by several chemotherapeutics, anti-cancer small molecule inhibitors, and ionizing radiation. Moreover, c-FLIP plays major roles in aiding the survival of immunosuppressive tumor-promoting immune cells and functions in inflammation, Alzheimer's disease (AD), and chronic obstructive pulmonary disease (COPD). Therefore, c-FLIP can serve as a versatile biomarker for cancer prognosis, a diagnostic marker for several diseases, and an effective therapeutic target. In this article, we review the functions of c-FLIP as an anti-apoptotic protein and negative prognostic factor in human cancers, and its roles in resistance to anticancer drugs, necroptosis and autophagy, immunosuppression, Alzheimer's disease, and COPD.
Collapse
Affiliation(s)
- Ahmad R Safa
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, USA
| | - Krzysztof Kamocki
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, USA
| | - M Reza Saadatzadeh
- Department of Neurosurgery, Indiana University School of Medicine, Indianapolis, USA
| | | |
Collapse
|
24
|
Qiu GH, Que W, Yan S, Zheng X, Xie X, Huang C, Yang X, Hooi SC. The pro-survival function of DLEC1 and its protection of cancer cells against 5-FU-induced apoptosis through up-regulation of BCL-XL. Cytotechnology 2019; 71:23-33. [PMID: 30607648 DOI: 10.1007/s10616-018-0258-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 09/15/2018] [Indexed: 02/06/2023] Open
Abstract
The tumor suppressor DLEC1 has been shown to promote cell proliferation when AP-2α2 is down-regulated in HCT116 stable clones, suggesting its pro-survival nature. However, the pro-survival function of DLEC1 has not been confirmed in other cells and its underlying mechanisms remain elusive. Therefore, we knocked down DLEC1 in a panel of cell lines and found that DLEC1 depletion caused various extents of cell death through intrinsic pathway. DLEC1 overexpression promoted cell survival and reduced cell death in cancer cells after 5-FU treatment, while DLEC1 down-regulation sensitized cancer cells to 5-FU. Further studies demonstrated that DLEC1 attenuated the increase in cleaved PARP, caspase-3 and caspase-7, the activity of caspase-9 and the diffusion of cytosolic cytochrome c from mitochondria. Our data also showed that BCL-XL was up-regulated by DLEC1 in stable clones after 5-FU treatment. Altogether, these results indicated that DLEC1 protects cells against cell death induced by 5-FU through the attenuation of active proteins in caspase cascade and the up-regulation of BCL-XL. Therefore, DLEC1 can be a pro-survival protein under certain circumstances and a potential therapeutic target for increasing sensitivity of cancer cells to 5-FU.
Collapse
Affiliation(s)
- Guo-Hua Qiu
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Longyan University, Longyan, 364012, Fujian, People's Republic of China.
- Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Fujian Province Universities, Longyan University, Longyan, 364012, Fujian, People's Republic of China.
- College of Life Sciences, Longyan University, Longyan, 364012, Fujian, People's Republic of China.
- Department of Physiology, Faculty of Medicine, National University of Singapore, Singapore, 117597, Singapore.
| | - Wutang Que
- Orthopedics Department, Longyan First Hospital, Longyan, 364000, Fujian, People's Republic of China
| | - Shanying Yan
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Longyan University, Longyan, 364012, Fujian, People's Republic of China
- Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Fujian Province Universities, Longyan University, Longyan, 364012, Fujian, People's Republic of China
- College of Life Sciences, Longyan University, Longyan, 364012, Fujian, People's Republic of China
| | - Xintian Zheng
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Longyan University, Longyan, 364012, Fujian, People's Republic of China
- Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Fujian Province Universities, Longyan University, Longyan, 364012, Fujian, People's Republic of China
- College of Life Sciences, Longyan University, Longyan, 364012, Fujian, People's Republic of China
| | - Xiaojin Xie
- Department of Physiology, Faculty of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Cuiqin Huang
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Longyan University, Longyan, 364012, Fujian, People's Republic of China
- Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Fujian Province Universities, Longyan University, Longyan, 364012, Fujian, People's Republic of China
- College of Life Sciences, Longyan University, Longyan, 364012, Fujian, People's Republic of China
| | - Xiaoyan Yang
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Longyan University, Longyan, 364012, Fujian, People's Republic of China
- Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Fujian Province Universities, Longyan University, Longyan, 364012, Fujian, People's Republic of China
- College of Life Sciences, Longyan University, Longyan, 364012, Fujian, People's Republic of China
| | - Shing Chuan Hooi
- Department of Physiology, Faculty of Medicine, National University of Singapore, Singapore, 117597, Singapore.
| |
Collapse
|
25
|
Fiore A, Ugel S, De Sanctis F, Sandri S, Fracasso G, Trovato R, Sartoris S, Solito S, Mandruzzato S, Vascotto F, Hippen KL, Mondanelli G, Grohmann U, Piro G, Carbone C, Melisi D, Lawlor RT, Scarpa A, Lamolinara A, Iezzi M, Fassan M, Bicciato S, Blazar BR, Sahin U, Murray PJ, Bronte V. Induction of immunosuppressive functions and NF-κB by FLIP in monocytes. Nat Commun 2018; 9:5193. [PMID: 30518925 PMCID: PMC6281604 DOI: 10.1038/s41467-018-07654-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/15/2018] [Indexed: 12/19/2022] Open
Abstract
Immunosuppression is a hallmark of tumor progression, and treatments that inhibit or deplete monocytic myeloid-derived suppressive cells could promote anti-tumor immunity. c-FLIP is a central regulator of caspase-8-mediated apoptosis and necroptosis. Here we show that low-dose cytotoxic chemotherapy agents cause apoptosis linked to c-FLIP down-regulation selectively in monocytes. Enforced expression of c-FLIP or viral FLIP rescues monocytes from cytotoxicity and concurrently induces potent immunosuppressive activity, in T cell cultures and in vivo models of tumor progression and immunotherapy. FLIP-transduced human blood monocytes can suppress graft versus host disease. Neither expression of FLIP in granulocytes nor expression of other anti-apoptotic genes in monocytes conferred immunosuppression, suggesting that FLIP effects on immunosuppression are specific to monocytic lineage and distinct from death inhibition. Mechanistically, FLIP controls a broad transcriptional program, partially by NF-κB activation. Therefore, modulation of FLIP in monocytes offers a means to elicit or block immunosuppressive myeloid cells.
Collapse
Affiliation(s)
- Alessandra Fiore
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy
- Max Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Stefano Ugel
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy.
| | - Francesco De Sanctis
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy
| | - Sara Sandri
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy
| | - Giulio Fracasso
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy
| | - Rosalinda Trovato
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy
| | - Silvia Sartoris
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy
| | - Samantha Solito
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, Padova, 35124, Italy
| | - Susanna Mandruzzato
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, Padova, 35124, Italy
- Istituto Oncologico Veneto IOV-IRCCS, Padova, 35124, Italy
| | - Fulvia Vascotto
- TRON-Translational Oncology, University Medical Center of Johannes Gutenberg University, Mainz, 55131, Germany
| | - Keli L Hippen
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, 55455, MN, USA
| | - Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Geny Piro
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, 37134, Italy
- Department of Medicine, Laboratory of Oncology and Molecular Therapy, University of Verona, Verona, 37134, Italy
| | - Carmine Carbone
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, 37134, Italy
| | - Davide Melisi
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, 37134, Italy
| | - Rita T Lawlor
- ARC-Net Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, 37134, Italy
| | - Aldo Scarpa
- ARC-Net Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, 37134, Italy
- Department of Pathology and Diagnostics, University of Verona, Verona, 37134, Italy
| | - Alessia Lamolinara
- Department of Medicine and Aging Science, Center of Excellence on Aging and Translational Medicine (CeSi-Met), University G. D'Annunzio of Chieti-Pescara, Chieti, 66100, Italy
| | - Manuela Iezzi
- Department of Medicine and Aging Science, Center of Excellence on Aging and Translational Medicine (CeSi-Met), University G. D'Annunzio of Chieti-Pescara, Chieti, 66100, Italy
| | - Matteo Fassan
- Department of Medicine-DIMED, University of Padova, Padova, 35124, Italy
| | - Silvio Bicciato
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, Modena, 41100, Italy
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, 55455, MN, USA
| | - Ugur Sahin
- TRON-Translational Oncology, University Medical Center of Johannes Gutenberg University, Mainz, 55131, Germany
- University Medical Center of the Johannes Gutenberg University, Mainz, 55131, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, 55131, Germany
| | - Peter J Murray
- Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.
| | - Vincenzo Bronte
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy.
| |
Collapse
|
26
|
Cytoplasmic FLIP(S) and nuclear FLIP(L) mediate resistance of castrate-resistant prostate cancer to apoptosis induced by IAP antagonists. Cell Death Dis 2018; 9:1081. [PMID: 30349042 PMCID: PMC6197283 DOI: 10.1038/s41419-018-1125-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/19/2018] [Accepted: 10/03/2018] [Indexed: 02/06/2023]
Abstract
Expression of tumor necrosis factor-α (TNFα) in the serum of prostate cancer patients is associated with poorer outcome and progression to castrate-resistant (CRPC) disease. TNFα promotes the activity of NFκB, which regulates a number of anti-apoptotic and proinflammatory genes, including those encoding the inhibitor of apoptosis proteins (IAPs); however, in the presence of IAP antagonists, TNFα can induce cell death. In the presence of recombinant or macrophage-derived TNFα, we found that IAP antagonists triggered degradation of cIAP1 and induced formation of Complex-IIb, consisting of caspase-8, FADD and RIPK1 in CRPC models; however, no, or modest levels of apoptosis were induced. This resistance was found to be mediated by both the long (L) and short (S) splice forms of the caspase-8 inhibitor, FLIP, another NFκB-regulated protein frequently overexpressed in CRPC. By decreasing FLIP expression at the post-transcriptional level in PC3 and DU145 cells (but not VCaP), the Class-I histone deacetylase (HDAC) inhibitor Entinostat promoted IAP antagonist-induced cell death in these models in a manner dependent on RIPK1, FADD and Caspase-8. Of note, Entinostat primarily targeted the nuclear rather than cytoplasmic pool of FLIP(L). While the cytoplasmic pool of FLIP(L) was highly stable, the nuclear pool was more labile and regulated by the Class-I HDAC target Ku70, which we have previously shown regulates FLIP stability. The efficacy of IAP antagonist (TL32711) and Entinostat combination and their effects on cIAP1 and FLIP respectively were confirmed in vivo, highlighting the therapeutic potential for targeting IAPs and FLIP in proinflammatory CRPC.
Collapse
|
27
|
Multiple Functions of Cellular FLIP Are Essential for Replication of Hepatitis B Virus. J Virol 2018; 92:JVI.00339-18. [PMID: 29875248 DOI: 10.1128/jvi.00339-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/24/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) infection is a leading cause of liver diseases; however, the host factors which facilitate the replication and persistence of HBV are largely unidentified. Cellular FLICE inhibitory protein (c-FLIP) is a typical antiapoptotic protein. In many cases of liver diseases, the expression level of c-FLIP is altered, which affects the fate of hepatocytes. We previously found that c-FLIP and its cleaved form interact with HBV X protein (HBx), which is essential for HBV replication, and regulate diverse cellular signals. In this study, we investigated the role of endogenous c-FLIP in HBV replication and its underlying mechanisms. The knockdown of endogenous c-FLIP revealed that this protein regulates HBV replication through two different mechanisms. (i) c-FLIP interacts with HBx and protects it from ubiquitin-dependent degradation. The N-terminal DED1 domain of c-FLIP is required for HBx stabilization. (ii) c-FLIP regulates the expression or stability of hepatocyte nuclear factors (HNFs), which have critical roles in HBV transcription and maintenance of hepatocytes. c-FLIP regulates the stability of HNFs through physical interactions. We verified our findings in three HBV infection systems: HepG2-NTCP cells, differentiated HepaRG cells, and primary human hepatocytes. In conclusion, our results identify c-FLIP as an essential factor in HBV replication. c-FLIP regulates viral replication through its multiple effects on viral and host proteins that have critical roles in HBV replication.IMPORTANCE Although the chronic hepatitis B virus (HBV) infection still poses a major health concern, the host factors which are required for the replication of HBV are largely uncharacterized. Our studies identify cellular FLICE inhibitory protein (c-FLIP) as an essential factor in HBV replication. We found the dual roles of c-FLIP in regulation of HBV replication: c-FLIP interacts with HBx and enhances its stability and regulates the expression or stability of hepatocyte nuclear factors which are essential for transcription of HBV genome. Our findings may provide a new target for intervention in persistent HBV infection.
Collapse
|
28
|
Gupta B, Ruttala HB, Poudel BK, Pathak S, Regmi S, Gautam M, Poudel K, Sung MH, Ou W, Jin SG, Jeong JH, Ku SK, Choi HG, Yong CS, Kim JO. Polyamino Acid Layer-by-Layer (LbL) Constructed Silica-Supported Mesoporous Titania Nanocarriers for Stimuli-Responsive Delivery of microRNA 708 and Paclitaxel for Combined Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2018; 10:24392-24405. [PMID: 29978708 DOI: 10.1021/acsami.8b06642] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cellular Fas-associated protein with death domain-like interleukin-1β-converting enzyme-inhibitory protein (c-FLIP), often strongly expressed in numerous cancers, plays a pivotal role in thwarting apoptosis and inducing chemotherapy resistance in cancer. An integrated approach combining chemotherapy with suppression of c-FLIP levels could prove paramount in the treatment of cancers with c-FLIP overexpression. In this study, we utilized a polymeric layer-by-layer (LbL) assembly of silica-supported mesoporous titania nanoparticles (MTNst) to co-deliver paclitaxel (PTX) and microRNA 708 (miR708) for simultaneous chemotherapy and c-FLIP suppression in colorectal carcinoma. The resulting LbL miR708/PTX-MTNst showed dose-dependent cytotoxicity in HCT-116 and DLD-1 colorectal carcinoma cell lines, which was remarkably superior to that of free PTX or LbL PTX-MTNst. LbL miR708/PTX-MTNst strongly inhibited c-FLIP expression and resulted in increased expression of proapoptotic proteins. In DLD-1 xenograft tumor-bearing mice, the nanoparticles accumulated in the tumor, resulting in remarkable tumor regression, with the PTX and miR708-loaded nanoparticles showing significantly greater inhibitory effects than the free PTX or PTX-loaded nanoparticles. Immunohistochemical analyses of the tumors further confirmed the remarkable apoptotic and antiproliferative effects of the nanoparticles, whereas organ histology reinforced the biocompatibility of the system. Therefore, the LbL miR708/PTX-MTNst system, owing to its ability to deliver both chemotherapeutic drug and inhibitory miRNA to the tumor site, shows great potential to treat colorectal carcinoma in clinical settings.
Collapse
Affiliation(s)
- Biki Gupta
- College of Pharmacy , Yeungnam University , 214-1, Dae-Dong , Gyeongsan 712-749 , Republic of Korea
| | - Hima Bindu Ruttala
- College of Pharmacy , Yeungnam University , 214-1, Dae-Dong , Gyeongsan 712-749 , Republic of Korea
| | - Bijay Kumar Poudel
- College of Pharmacy , Yeungnam University , 214-1, Dae-Dong , Gyeongsan 712-749 , Republic of Korea
| | - Shiva Pathak
- College of Pharmacy , Yeungnam University , 214-1, Dae-Dong , Gyeongsan 712-749 , Republic of Korea
| | - Shobha Regmi
- College of Pharmacy , Yeungnam University , 214-1, Dae-Dong , Gyeongsan 712-749 , Republic of Korea
| | - Milan Gautam
- College of Pharmacy , Yeungnam University , 214-1, Dae-Dong , Gyeongsan 712-749 , Republic of Korea
| | - Kishwor Poudel
- College of Pharmacy , Yeungnam University , 214-1, Dae-Dong , Gyeongsan 712-749 , Republic of Korea
| | - Min Hyun Sung
- College of Pharmacy , Yeungnam University , 214-1, Dae-Dong , Gyeongsan 712-749 , Republic of Korea
| | - Wenquan Ou
- College of Pharmacy , Yeungnam University , 214-1, Dae-Dong , Gyeongsan 712-749 , Republic of Korea
| | - Sung Giu Jin
- Department of Pharmaceutical Engineering , Dankook University , 119 Dandae-ro , Dongnam-gu, Cheonan 31116 , Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy , Yeungnam University , 214-1, Dae-Dong , Gyeongsan 712-749 , Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine , Daegu Haany University , Gyeongsan 712-702 , Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology , Hanyang University , 55, Hanyangdaehak-ro , Sangnok-gu, Ansan 426-791 , Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy , Yeungnam University , 214-1, Dae-Dong , Gyeongsan 712-749 , Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy , Yeungnam University , 214-1, Dae-Dong , Gyeongsan 712-749 , Republic of Korea
| |
Collapse
|
29
|
Humphreys L, Espona-Fiedler M, Longley DB. FLIP as a therapeutic target in cancer. FEBS J 2018; 285:4104-4123. [PMID: 29806737 DOI: 10.1111/febs.14523] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/11/2018] [Accepted: 05/24/2018] [Indexed: 12/13/2022]
Abstract
One of the classic hallmarks of cancer is disruption of cell death signalling. Inhibition of cell death promotes tumour growth and metastasis, causes resistance to chemo- and radiotherapies as well as targeted agents, and is frequently due to overexpression of antiapoptotic proteins rather than loss of pro-apoptotic effectors. FLIP is a major apoptosis-regulatory protein frequently overexpressed in solid and haematological cancers, in which its high expression is often correlated with poor prognosis. FLIP, which is expressed as long (FLIP(L)) and short (FLIP(S)) splice forms, achieves its cell death regulatory functions by binding to FADD, a critical adaptor protein which links FLIP to the apical caspase in the extrinsic apoptotic pathway, caspase-8, in a number of cell death regulating complexes, such as the death-inducing signalling complexes (DISCs) formed by death receptors. FLIP also plays a key role (together with caspase-8) in regulating another form of cell death termed programmed necrosis or 'necroptosis', as well as in other key cellular processes that impact cell survival, including autophagy. In addition, FLIP impacts activation of the intrinsic mitochondrial-mediated apoptotic pathway by regulating caspase-8-mediated activation of the pro-apoptotic Bcl-2 family member Bid. It has been demonstrated that FLIP can not only inhibit death receptor-mediated apoptosis, but also cell death induced by a range of clinically relevant chemotherapeutic and targeted agents as well as ionizing radiation. More recently, key roles for FLIP in promoting the survival of immunosuppressive tumour-promoting immune cells have been discovered. Thus, FLIP is of significant interest as an anticancer therapeutic target. In this article, we review FLIP's biology and potential ways of targeting this important tumour and immune cell death regulator.
Collapse
Affiliation(s)
- Luke Humphreys
- Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| | - Margarita Espona-Fiedler
- Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| | - Daniel B Longley
- Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| |
Collapse
|
30
|
Kim EA, Kim SW, Nam J, Sung EG, Song IH, Kim JY, Kwon TK, Lee TJ. Inhibition of c-FLIPL expression by miRNA-708 increases the sensitivity of renal cancer cells to anti-cancer drugs. Oncotarget 2017; 7:31832-46. [PMID: 27092874 PMCID: PMC5077980 DOI: 10.18632/oncotarget.7149] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 01/23/2016] [Indexed: 12/23/2022] Open
Abstract
Dysregulation of the anti-apoptotic protein, cellular FLICE-like inhibitory protein (c-FLIP), has been associated with tumorigenesis and chemoresistance in various human cancers. Therefore, c-FLIP is an excellent target for therapeutic intervention. MicroRNAs (miRNAs) are small non-coding RNAs that are involved in tumorigenesis, tumor suppression, and resistance or sensitivity to anti-cancer drugs. However, whether miRNAs can suppress c-FLIPL expression in cancer cells is unclear. The aim of this study was to identify miRNAs that could inhibit the growth of renal cancer cells and induce cell death by inhibiting c-FLIPL expression. We found that MiRNA-708 and c-FLIPL expression were inversely correlated. While c-FLIPL expression was upregulated, miRNA-708 was rarely expressed in renal cancer cells. Luciferase reporter assays demonstrated that miRNA-708 negatively regulated c-FLIPL expression by binding to the miRNA-708 binding site in the 3' untranslated region (3'UTR) of c-FLIPL. Ectopic expression of miRNA-708 increased the accumulation of sub-G1 populations and cleavage of procaspase-3 and PARP, which could be prevented by pretreatment with the pan-caspase inhibitor, Z-VAD. Ectopic expression of miRNA-708 also increased the sensitivity to various apoptotic stimuli such as tumor necrosis factor-related apoptosis-inducing ligand, doxorubicin (Dox), and thapsigargin in Caki cells. Interestingly, miRNA-708 specifically repressed c-FLIPL without any change in c-FLIPs expression. In contrast, inhibition of endogenous miRNA-708 using antago-miRNAs resulted in an increase in c-FLIPL protein expression. The expression of c-FLIPL was upregulated in renal cell carcinoma (RCC) tissues compared to normal tissues. In contrast, miRNA-708 expression was reduced in RCC tissues. Finally, miRNA-708 enhanced the tumor-suppressive effect of Dox in a xenograft model of human RCC. In conclusion, miRNA-708 acts as a tumor suppressor because it negatively regulates the anti-apoptotic protein c-FLIPL and regulates the sensitivity of renal cancer cells to various apoptotic stimuli.
Collapse
Affiliation(s)
- Eun-Ae Kim
- Department of Anatomy, College of Medicine, Yeungnam University, Nam-gu, Daegu, Republic of Korea
| | - Sang-Woo Kim
- Department of Biological Sciences, Pusan National University, Busan, Republic of Korea
| | - Jehyun Nam
- Department of Biological Sciences, Pusan National University, Busan, Republic of Korea
| | - Eon-Gi Sung
- Department of Anatomy, College of Medicine, Yeungnam University, Nam-gu, Daegu, Republic of Korea
| | - In-Hwan Song
- Department of Anatomy, College of Medicine, Yeungnam University, Nam-gu, Daegu, Republic of Korea
| | - Joo-Young Kim
- Department of Anatomy, College of Medicine, Yeungnam University, Nam-gu, Daegu, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Dalseo-Gu, Daegu, Republic of Korea
| | - Tae-Jin Lee
- Department of Anatomy, College of Medicine, Yeungnam University, Nam-gu, Daegu, Republic of Korea
| |
Collapse
|
31
|
Hutchinson RA, Coleman HG, Gately K, Young V, Nicholson S, Cummins R, Kay E, Hynes SO, Dunne PD, Senevirathne S, Hamilton PW, McArt DG, Longley DB. IHC-based subcellular quantification provides new insights into prognostic relevance of FLIP and procaspase-8 in non-small-cell lung cancer. Cell Death Discov 2017; 3:17050. [PMID: 28904817 PMCID: PMC5594421 DOI: 10.1038/cddiscovery.2017.50] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 05/24/2017] [Accepted: 05/25/2017] [Indexed: 12/23/2022] Open
Abstract
In this study, we developed an image analysis algorithm for quantification of two potential apoptotic biomarkers in non-small-cell lung cancer (NSCLC): FLIP and procaspase-8. Immunohistochemical expression of FLIP and procaspase-8 in 184 NSCLC tumors were assessed. Individual patient cores were segmented and classified as tumor and stroma using the Definiens Tissue Studio. Subsequently, chromogenic expression of each biomarker was measured separately in the nucleus and cytoplasm and reported as a quantitative histological score. The software package pROC was applied to define biomarker thresholds. Cox proportional hazards analysis was applied to generate hazard ratios (HRs) and associated 95% CI for survival. High cytoplasmic expression of tumoral (but not stromal) FLIP was associated with a 2.5-fold increased risk of death in lung adenocarcinoma patients, even when adjusted for known confounders (HR 2.47, 95% CI 1.14-5.35). Neither nuclear nor cytoplasmic tumoral procaspase-8 expression was associated with overall survival in lung adenocarcinoma patients; however, there was a significant trend (P for trend=0.03) for patients with adenocarcinomas with both high cytoplasmic FLIP and high cytoplasmic procaspase-8 to have a multiplicative increased risk of death. Notably, high stromal nuclear procaspase-8 expression was associated with a reduced risk of death in lung adenocarcinoma patients (adjusted HR 0.31, 95% CI 0.15-0.66). On further examination, the cells with high nuclear procaspase-8 were found to be of lymphoid origin, suggesting that the better prognosis of patients with tumors with high stromal nuclear procaspase-8 is related to immune infiltration, a known favorable prognostic factor. No significant associations were detected in analysis of lung squamous cell carcinoma patients. Our results suggest that cytoplasmic expression of FLIP in the tumor and nuclear expression of procaspase-8 in the stroma are prognostically relevant in non-small-cell adenocarcinomas but not in squamous cell carcinomas of the lung.
Collapse
Affiliation(s)
- Ryan A Hutchinson
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland, UK
| | - Helen G Coleman
- Centre for Public Health, Queen’s University Belfast, Belfast, Northern Ireland, UK
| | - Kathy Gately
- Department of Cardiothoracic Surgery, St James’s Hospital, Dublin, Ireland
| | - Vincent Young
- Department of Cardiothoracic Surgery, St James’s Hospital, Dublin, Ireland
| | - Siobhan Nicholson
- Department of Cardiothoracic Surgery, St James’s Hospital, Dublin, Ireland
| | - Robert Cummins
- Department of Pathology, Education and Research Centre, Royal College of Surgeons of Ireland, Beaumont Hospital, Dublin, Ireland
| | - Elaine Kay
- Department of Pathology, Education and Research Centre, Royal College of Surgeons of Ireland, Beaumont Hospital, Dublin, Ireland
| | - Sean O Hynes
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland, UK
| | - Philip D Dunne
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland, UK
| | - Seedevi Senevirathne
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland, UK
| | - Peter W Hamilton
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland, UK
| | - Darragh G McArt
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland, UK
| | - Daniel B Longley
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland, UK
| |
Collapse
|
32
|
Kong W, Mou X, Deng J, Di B, Zhong R, Wang S, Yang Y, Zeng W. Differences of immune disorders between Alzheimer's disease and breast cancer based on transcriptional regulation. PLoS One 2017; 12:e0180337. [PMID: 28719625 PMCID: PMC5515412 DOI: 10.1371/journal.pone.0180337] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 06/14/2017] [Indexed: 01/01/2023] Open
Abstract
Although chronic inflammation and immune disorders are of great importance to the pathogenesis of both dementia and cancer, the pathophysiological mechanisms are not clearly understood. In recent years, growing epidemiological evidence and meta-analysis data suggest an inverse association between Alzheimer's disease (AD), which is the most common form of dementia, and cancer. It has been revealed that some common genes and biological processes play opposite roles in AD and cancer; however, the biological immune mechanism for the inverse association is not clearly defined. An unsupervised matrix decomposition two-stage bioinformatics procedure was adopted to investigate the opposite behaviors of the immune response in AD and breast cancer (BC) and to discover the underlying transcriptional regulatory mechanisms. Fast independent component analysis (FastICA) was applied to extract significant genes from AD and BC microarray gene expression data. Based on the extracted data, the shared transcription factors (TFs) from AD and BC were captured. Second, the network component analysis (NCA) algorithm in this study was presented to quantitatively deduce the TF activities and regulatory influences because quantitative dynamic regulatory information for TFs is not available via microarray techniques. Based on the NCA results and reconstructed transcriptional regulatory networks, inverse regulatory processes and some known innate immune responses were described in detail. Many of the shared TFs and their regulatory processes were found to be closely related to the adaptive immune response from dramatically different directions and to play crucial roles in both AD and BC pathogenesis. From the above findings, the opposing cellular behaviors demonstrate an invaluable opportunity to gain insights into the pathogenesis of these two types of diseases and to aid in developing new treatments.
Collapse
Affiliation(s)
- Wei Kong
- College of Information Engineering, Shanghai Maritime University, Haigang Ave., Shanghai, P. R. China
| | - Xiaoyang Mou
- Department of Biochemistry, Rowan University and Guava Medicine, Glassboro, New Jersey, United States of America
| | - Jin Deng
- College of Information Engineering, Shanghai Maritime University, Haigang Ave., Shanghai, P. R. China
| | - Benteng Di
- College of Information Engineering, Shanghai Maritime University, Haigang Ave., Shanghai, P. R. China
| | - Ruxing Zhong
- College of Information Engineering, Shanghai Maritime University, Haigang Ave., Shanghai, P. R. China
| | - Shuaiqun Wang
- College of Information Engineering, Shanghai Maritime University, Haigang Ave., Shanghai, P. R. China
| | - Yang Yang
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Weiming Zeng
- College of Information Engineering, Shanghai Maritime University, Haigang Ave., Shanghai, P. R. China
| |
Collapse
|
33
|
Shen L, Sun Z, Zhao F, Wang W, Zhang W, Zhu H. Expression of c-FLIP in a rat model of sepsis and its effects on endothelial apoptosis. Mol Med Rep 2017; 16:231-237. [PMID: 28498469 PMCID: PMC5482130 DOI: 10.3892/mmr.2017.6564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 02/15/2017] [Indexed: 11/06/2022] Open
Abstract
Sepsis is characterized by the impaired regulation of inflammatory responses. Apoptosis is important in the pathogenesis of sepsis. Cellular FLICE-inhibitory protein (c-FLIP) is a catalytically inactive caspase-8 homologue, which negatively interferes with apoptotic signaling. The role of c-FLIP in sepsis and in endothelial cell apoptosis, a critical step in the pathogenesis of sepsis, remains controversial. In the present study, to investigate the relationship between c-FLIP and sepsis, a rat model of sepsis was induced by cecal ligation and puncture, and western blot analysis was used to detect the expression of c-FLIPL, the long isoform of c-FLIP. Lower protein expression levels of c-FLIPL were found in the brain, intestine and lung of the rat sepsis model, compared with the rats in the sham surgery group. The association between the expression of c-FLIPL and endothelial cell apoptosis was further examined in vitro by c-FLIPL overexpression and flow cytometry, which demonstrated that the expression of c-FLIPL was inversely correlated with endothelial cell apoptosis. These data suggested that c-FLIP may be important in sepsis and shed light on therapeutic strategies.
Collapse
Affiliation(s)
- Lei Shen
- Department of Intensive Care Unit, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Zhengda Sun
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143, USA
| | - Feng Zhao
- Department of Intensive Care Unit, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Wei Wang
- Department of Intensive Care Unit, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Wenhong Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Hechen Zhu
- Department of Intensive Care Unit, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
34
|
Autocrine and Paracrine Mechanisms Promoting Chemoresistance in Cholangiocarcinoma. Int J Mol Sci 2017; 18:ijms18010149. [PMID: 28098760 PMCID: PMC5297782 DOI: 10.3390/ijms18010149] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/19/2016] [Accepted: 01/06/2017] [Indexed: 02/07/2023] Open
Abstract
Resistance to conventional chemotherapeutic agents, a typical feature of cholangiocarcinoma, prevents the efficacy of the therapeutic arsenal usually used to combat malignancy in humans. Mechanisms of chemoresistance by neoplastic cholangiocytes include evasion of drug-induced apoptosis mediated by autocrine and paracrine cues released in the tumor microenvironment. Here, recent evidence regarding molecular mechanisms of chemoresistance is reviewed, as well as associations between well-developed chemoresistance and activation of the cancer stem cell compartment. It is concluded that improved understanding of the complex interplay between apoptosis signaling and the promotion of cell survival represent potentially productive areas for active investigation, with the ultimate aim of encouraging future studies to unveil new, effective strategies able to overcome current limitations on treatment.
Collapse
|
35
|
Zhang J, Jiang HY, Zhang LK, Xu WL, Qiao YT, Zhu XG, Liu W, Zheng QQ, Hua ZC. C-FLIP L Modulated Wnt/β-Catenin Activation via Association with TIP49 Protein. J Biol Chem 2016; 292:2132-2142. [PMID: 28028178 DOI: 10.1074/jbc.m116.753251] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 12/05/2016] [Indexed: 12/24/2022] Open
Abstract
Cellular FLICE-like inhibitory protein (c-FLIPL) is a key inhibitory protein in the extrinsic apoptotic pathway. Recent studies showed that c-FLIPL could translocate into the nucleus and might be involved in the Wnt signaling pathway. The nuclear function of c-FLIPL was still unclear. Here we found a novel c-FLIPL-associated protein TIP49, which is a nuclear protein identified as a cofactor in the transcriptional regulation of β-catenin. They had co-localization in the nucleus and the DED domain of c-FLIPL was required for the association with TIP49. By performing ChIP experiments, C-FLIPL was detected in the ITF-2 locus and facilitated TIP49 accumulation in the formation of complexes at the T-cell-specific transcription factor site of human ITF-2 promoter. When TIP49 knockdown, c-FLIPL-driven Wnt activation, and cell proliferation were inhibited, suggesting that a role of nuclear c-FLIPL involved in modulation of the Wnt pathway was in a TIP49-dependent manner. Elevated expression of c-FLIPL and TIP49 that coincided in human lung cancers were analyzed in silico using the Oncomine database. Their high expressions were reconfirmed in six lung cancer cell lines and correlated with cell growth. The association of c-FLIPL and TIP49 provided an additional mechanism involved in c-FLIPL-mediated functions, including Wnt activation.
Collapse
Affiliation(s)
- Jing Zhang
- From The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu and .,the Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou 213164, Jiangsu, People's Republic of China
| | - Heng-Yi Jiang
- From The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu and
| | - Lin-Kai Zhang
- From The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu and
| | - Wen-Ling Xu
- From The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu and
| | - Yi-Ting Qiao
- From The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu and
| | - Xu-Guo Zhu
- From The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu and
| | - Wan Liu
- From The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu and
| | - Qian-Qian Zheng
- From The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu and
| | - Zi-Chun Hua
- From The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu and .,the Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou 213164, Jiangsu, People's Republic of China
| |
Collapse
|
36
|
Wen C, Chen J, Zhang D, Wang H, Che J, Qin Q, He L, Cai Z, Lin M, Lou Q, Huang L, Chen D, Iwamoto A, Ren D, Wang L, Lan P, Wang J, Liu H, Yang X. Pseudolaric acid B induces mitotic arrest and apoptosis in both 5-fluorouracil-sensitive and -resistant colorectal cancer cells. Cancer Lett 2016; 383:295-308. [PMID: 27713084 DOI: 10.1016/j.canlet.2016.09.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/18/2016] [Accepted: 09/18/2016] [Indexed: 01/10/2023]
Abstract
5-fluorouracil (5-FU)-based chemotherapy is the main chemotherapeutic approach for colorectal cancer (CRC) treatment. Because chemoresistance occurs frequently and significantly limits CRC therapies, a novel agent is needed. Pseudolaric acid B (PAB), a small molecule derived from the Chinese medicinal herb ''Tujinpi'', exhibits strong cytotoxic effects on a variety of cancers. However, the detailed mechanisms by which PAB inhibits CRC cell growth and its potential role in overcoming 5-FU resistance have not been well studied. In this study, we showed that PAB significantly inhibited the viability of various CRC cell lines but induced minor cytotoxicity in normal cells. Both the in vitro and in vivo results showed that PAB induced proliferation inhibition, mitotic arrest and subsequently caspase-dependent apoptosis in both 5-FU-sensitive and -resistant CRC cells. Moreover, PAB was shown to interfere with CRC cell mitotic spindle apparatus and activate the spindle assembly checkpoint. Finally, CDK1 activity was involved in PAB-induced mitotic arrest and apoptosis in CRC cells. Taken together, these data reveal that PAB induces CRC cell mitotic arrest followed by apoptosis and overcomes 5-FU resistance in vitro and in vivo, suggesting that PAB may be a potential agent for CRC treatment, particularly for 5-FU-resistant CRC.
Collapse
Affiliation(s)
- Chuangyu Wen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Junxiong Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Di Zhang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huihui Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Jia Che
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Qiyuan Qin
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lu He
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zerong Cai
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mengmeng Lin
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiong Lou
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Lanlan Huang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Daici Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Aikichi Iwamoto
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Donglin Ren
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ping Lan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianping Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huanliang Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| | - Xiangling Yang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
37
|
Kim JH, Kim MJ, Choi KC, Son J. Quercetin sensitizes pancreatic cancer cells to TRAIL-induced apoptosis through JNK-mediated cFLIP turnover. Int J Biochem Cell Biol 2016; 78:327-334. [DOI: 10.1016/j.biocel.2016.07.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 07/27/2016] [Indexed: 10/21/2022]
|
38
|
McLaughlin KA, Nemeth Z, Bradley CA, Humphreys L, Stasik I, Fenning C, Majkut J, Higgins C, Crawford N, Holohan C, Johnston PG, Harrison T, Hanna GG, Butterworth KT, Prise KM, Longley DB. FLIP: A Targetable Mediator of Resistance to Radiation in Non–Small Cell Lung Cancer. Mol Cancer Ther 2016; 15:2432-2441. [DOI: 10.1158/1535-7163.mct-16-0211] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/06/2016] [Indexed: 11/16/2022]
|
39
|
Goldar S, Khaniani MS, Derakhshan SM, Baradaran B. Molecular mechanisms of apoptosis and roles in cancer development and treatment. Asian Pac J Cancer Prev 2016; 16:2129-44. [PMID: 25824729 DOI: 10.7314/apjcp.2015.16.6.2129] [Citation(s) in RCA: 396] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Programmed cell death (PCD) or apoptosis is a mechanism which is crucial for all multicellular organisms to control cell proliferation and maintain tissue homeostasis as well as eliminate harmful or unnecessary cells from an organism. Defects in the physiological mechanisms of apoptosis may contribute to different human diseases like cancer. Identification of the mechanisms of apoptosis and its effector proteins as well as the genes responsible for apoptosis has provided a new opportunity to discover and develop novel agents that can increase the sensitivity of cancer cells to undergo apoptosis or reset their apoptotic threshold. These novel targeted therapies include those targeting anti-apoptotic Bcl-2 family members, p53, the extrinsic pathway, FLICE-inhibitory protein (c-FLIP), inhibitor of apoptosis (IAP) proteins, and the caspases. In recent years a number of these novel agents have been assessed in preclinical and clinical trials. In this review, we introduce some of the key regulatory molecules that control the apoptotic pathways, extrinsic and intrinsic death receptors, discuss how defects in apoptotic pathways contribute to cancer, and list several agents being developed to target apoptosis.
Collapse
Affiliation(s)
- Samira Goldar
- Department of Biochemistry and Clinical Labratorary, Division of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran E-mail :
| | | | | | | |
Collapse
|
40
|
Todoroki K, Ogasawara S, Akiba J, Nakayama M, Naito Y, Seki N, Kusukawa J, Yano H. CD44v3+/CD24- cells possess cancer stem cell-like properties in human oral squamous cell carcinoma. Int J Oncol 2015; 48:99-109. [PMID: 26647656 PMCID: PMC4734600 DOI: 10.3892/ijo.2015.3261] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 11/06/2015] [Indexed: 12/27/2022] Open
Abstract
Cancer stem cells (CSCs) or cancer stem cell-like cells (CSC-LCs) are a minority population of cells that relate to tumor progression, metastasis and drug resistance. To identify CSC-LCs in oral squamous cell carcinoma (OSCC), we used two OSCC cell lines, SAS and OSC20, and cell surface markers, CD44v3 and CD24. In addition, we examined CD44v3 and CD24 expression immunohistochemically and evaluated the relationship between the expression and clinicopathological parameters in 50 OSCC tissues. In SAS and OSC20, CD44v3+/CD24- cells showed a higher sphere forming ability than the other fractions, i.e., CD44v3+/CD24+, CD44v3-/CD24- and CD44v3-/CD24+ cells. The proportion of CD44v3+/CD24- cells in SAS and OSC20 was 10.7 and 24.1%, respectively. Regarding SAS, CD44v3+/CD24- cells also showed a higher drug resistance for CDDP, 5-FU and cetuximab and expressed higher mRNA levels of CSC property-related genes than the other cell fractions. The tumorigenicity of CD44v3+/CD24- cells was not significantly different from the other fractions in SAS. An immunohistochemical study revealed a significant correlation between CD44v3 expression in the invasive portion and lymph node metastasis. Kaplan Meier analysis revealed cases with CD44v3 expression in the invasive portion tended to show poor overall survival (OS) compared with those without CD44v3, and there was a significant difference in OS between CD44v3+/CD24- and CD44v3-/CD24- immunophenotypes in the invasive portion. In conclusion, the results suggest that the CD44v3+/CD24- cell population displays CSC-LC properties in a human OSCC cell line. Additionally, we present evidence that CD44v3 immunoexpression and CD44v3+/CD24- immunophenotypes could give prognostic information associated with unfavorable clinical outcomes.
Collapse
Affiliation(s)
- Keita Todoroki
- Department of Pathology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Sachiko Ogasawara
- Department of Pathology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Jun Akiba
- Department of Pathology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Masamichi Nakayama
- Department of Pathology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Yoshiki Naito
- Department of Pathology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Naoko Seki
- Dental and Oral Medical Center, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Jingo Kusukawa
- Dental and Oral Medical Center, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Hirohisa Yano
- Department of Pathology, Kurume University School of Medicine, Kurume 830-0011, Japan
| |
Collapse
|
41
|
Gaidos G, Panaitiu AE, Guo B, Pellegrini M, Mierke DF. Identification and Characterization of the Interaction Site between cFLIPL and Calmodulin. PLoS One 2015; 10:e0141692. [PMID: 26529318 PMCID: PMC4631386 DOI: 10.1371/journal.pone.0141692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/11/2015] [Indexed: 11/18/2022] Open
Abstract
Overexpression of the cellular FLICE-like inhibitory protein (cFLIP) has been reported in a number of tumor types. As an inactive procaspase-8 homologue, cFLIP is recruited to the intracellular assembly known as the Death Inducing Signaling Complex (DISC) where it inhibits apoptosis, leading to cancer cell proliferation. Here we characterize the molecular details of the interaction between cFLIPL and calmodulin, a ubiquitous calcium sensing protein. By expressing the individual domains of cFLIPL, we demonstrate that the interaction with calmodulin is mediated by the N-terminal death effector domain (DED1) of cFLIPL. Additionally, we mapped the interaction to a specific region of the C-terminus of DED1, referred to as DED1 R4. By designing DED1/DED2 chimeric constructs in which the homologous R4 regions of the two domains were swapped, calmodulin binding properties were transferred to DED2 and removed from DED1. Furthermore, we show that the isolated DED1 R4 peptide binds to calmodulin and solve the structure of the peptide-protein complex using NMR and computational refinement. Finally, we demonstrate an interaction between cFLIPL and calmodulin in cancer cell lysates. In summary, our data implicate calmodulin as a potential player in DISC-mediated apoptosis and provide evidence for a specific interaction with the DED1 of cFLIPL.
Collapse
Affiliation(s)
- Gabriel Gaidos
- Chemistry Department, Dartmouth College, Hanover, NH, United States of America
| | | | - Bingqian Guo
- Chemistry Department, Dartmouth College, Hanover, NH, United States of America
| | - Maria Pellegrini
- Chemistry Department, Dartmouth College, Hanover, NH, United States of America
| | - Dale F. Mierke
- Chemistry Department, Dartmouth College, Hanover, NH, United States of America
- * E-mail:
| |
Collapse
|
42
|
Carson R, Celtikci B, Fenning C, Javadi A, Crawford N, Carbonell LP, Lawler M, Longley DB, Johnston PG, Van Schaeybroeck S. HDAC Inhibition Overcomes Acute Resistance to MEK Inhibition in BRAF-Mutant Colorectal Cancer by Downregulation of c-FLIPL. Clin Cancer Res 2015; 21:3230-3240. [PMID: 25813020 PMCID: PMC4504978 DOI: 10.1158/1078-0432.ccr-14-2701] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/06/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Activating mutations in the BRAF oncogene are found in 8% to 15% of colorectal cancer patients and have been associated with poor survival. In contrast with BRAF-mutant (MT) melanoma, inhibition of the MAPK pathway is ineffective in the majority of BRAFMT colorectal cancer patients. Therefore, identification of novel therapies for BRAFMT colorectal cancer is urgently needed. EXPERIMENTAL DESIGN BRAFMT and wild-type (WT) colorectal cancer models were assessed in vitro and in vivo. Small-molecule inhibitors of MEK1/2, MET, and HDAC were used, overexpression and siRNA approaches were applied, and cell death was assessed by flow cytometry, Western blotting, cell viability, and caspase activity assays. RESULTS Increased c-MET-STAT3 signaling was identified as a novel adaptive resistance mechanism to MEK inhibitors (MEKi) in BRAFMT colorectal cancer models in vitro and in vivo. Moreover, MEKi treatment resulted in acute increases in transcription of the endogenous caspase-8 inhibitor c-FLIPL in BRAFMT cells, but not in BRAFWT cells, and inhibition of STAT3 activity abrogated MEKi-induced c-FLIPL expression. In addition, treatment with c-FLIP-specific siRNA or HDAC inhibitors abrogated MEKi-induced upregulation of c-FLIPL expression and resulted in significant increases in MEKi-induced cell death in BRAFMT colorectal cancer cells. Notably, combined HDAC inhibitor/MEKi treatment resulted in dramatically attenuated tumor growth in BRAFMT xenografts. CONCLUSIONS Our findings indicate that c-MET/STAT3-dependent upregulation of c-FLIPL expression is an important escape mechanism following MEKi treatment in BRAFMT colorectal cancer. Thus, combinations of MEKi with inhibitors of c-MET or c-FLIP (e.g., HDAC inhibitors) could be potential novel treatment strategies for BRAFMT colorectal cancer.
Collapse
Affiliation(s)
- Robbie Carson
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Basak Celtikci
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Cathy Fenning
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Arman Javadi
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Nyree Crawford
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Lucia Perez Carbonell
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Mark Lawler
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Daniel B. Longley
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Patrick G. Johnston
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Sandra Van Schaeybroeck
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| |
Collapse
|
43
|
Lee SW, Cho JM, Cho HJ, Kang JY, Kim EK, Yoo TK. Expression levels of heat shock protein 27 and cellular FLICE-like inhibitory protein in prostate cancer correlate with Gleason score sum and pathologic stage. Korean J Urol 2015; 56:505-14. [PMID: 26175869 PMCID: PMC4500807 DOI: 10.4111/kju.2015.56.7.505] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/17/2015] [Indexed: 01/26/2023] Open
Abstract
Purpose Heat shock protein (HSP) 27 protects the cell by controlling apoptosis and immune reactions, and c-FLIP (cellular-FLICE inhibitory protein) inhibits apoptosis by inhibiting caspase-8 activity. We investigated the relationship of HSP27 and c-FLIP expression to prostate-specific antigen, Gleason score sum (GSS), and pathologic stage. Materials and Methods Samples from 163 patients between May 2004 and April 2010 were analyzed: 83 from patients that had underwent a radical prostatectomy, and 80 from those that underwent transurethral resection of the prostate to alleviate urinary symptoms from benign prostate hyperplasia. c-FLIP and HSP27 expression were observed by immunohistochemistry staining. Samples with less than 5% expression-positive cells were scored as 1, with 5%-50% were scored as 2, and with more than 50% were scored as 3. Local reactions were identified as 0.5 and evaluated. Results Both the presence of HSP27 within the tumor and the number of cancer cells positive for HSP27 were significantly correlated to GSS and pathologic stage (p<0.001, p=0.001, p<0.001, p<0.001). The same was true for c-FLIP expression (p<0.001). GSS was more highly correlated to HSP27 expression than to c-FLIP expression (r=0.814 for HSP27, r=0.776 for c-FLIP), as was pathologic stage (r=0.592 for HSP27, r=0.554 for c-FLIP). Conclusions In prostate cancer, higher GSS and a more advanced pathologic stage were associated with a higher likelihood of having a HSP27-positive tumor and more HSP27-positive tumor cells. HSP27 expression was correlated with GSS and prostate cancer stage. A more advanced pathologic stage corresponded to a higher likelihood of having a c-FLIP-positive tumor and more c-FLIP-positive tumor cells. HSP27 expression had a higher correlation with prostate cancer stage and GSS than c-FLIP expression did.
Collapse
Affiliation(s)
- Seung Wook Lee
- Department of Urology, Hanyang University Guri Hospital, Hanyang University School of Medicine, Guri, Korea
| | - Jeoung Man Cho
- Department of Urology, Eulji University College of Medicine, Daejeon, Korea
| | - Hee Ju Cho
- Department of Urology, Eulji University College of Medicine, Daejeon, Korea
| | - Jung Yoon Kang
- Department of Urology, Eulji University College of Medicine, Daejeon, Korea
| | - Eun Kyung Kim
- Department of Pathology, Eulji University College of Medicine, Daejeon, Korea
| | - Tag Keun Yoo
- Department of Urology, Eulji University College of Medicine, Daejeon, Korea
| |
Collapse
|
44
|
Fulda S. Targeting apoptosis for anticancer therapy. Semin Cancer Biol 2015; 31:84-8. [DOI: 10.1016/j.semcancer.2014.05.002] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 05/10/2014] [Accepted: 05/13/2014] [Indexed: 11/16/2022]
|
45
|
Ullenhag GJ, Al-Attar A, Mukherjee A, Green AR, Ellis IO, Durrant LG. The TRAIL system is over-expressed in breast cancer and FLIP a marker of good prognosis. J Cancer Res Clin Oncol 2015; 141:505-14. [PMID: 25230899 DOI: 10.1007/s00432-014-1822-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 09/01/2014] [Indexed: 02/04/2023]
Abstract
BACKGROUND Breast cancer is the most common cancer in women. The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathway transmits apoptotic signals. Novel anticancer agents that activate this system are in clinical development, including anti-breast cancer. METHODS The tissue microarray technique was applied. We used an array of breast cancer tissues from a large group of patients (>800) to assess the protein expression of TRAIL-R1, TRAIL-R2, the long isoform of FLICE-inhibitory protein and total FLICE-inhibitory protein (FLIP(L) and FLIP(T)). Disease-free survival was examined by Kaplan-Meier estimates and the log-rank test. The independence of prognostic factors was determined by Cox multivariate analysis. RESULTS High intra-tumoral expression of all these proteins of the TRAIL pathway was found. The TRAIL receptors and FLIP(L) were not associated with survival. On univariate analysis, strong FLIP(T) expression was associated with a significantly better survival (p = 0.001). On multivariate analysis using the Cox proportional hazards model, FLIP(T) phenotype was significantly associated with a good prognosis in this series (HR 0.52, 95 % CI 0.35-0.78, p = 0.039). Results indicate that this association is valid for all the biological subtypes of breast cancer. The expression of FLIP(T) was especially high in the luminal subtype, known for its good prognosis. CONCLUSIONS These findings support the use of agonistic TRAIL antibodies and drugs targeting FLIP in breast cancer patients. Over-expression of FLIP(T) but not TRAIL-R1, TRAIL-R2 or FLIP(L) provides stage-independent prognostic information in breast cancer patients. This indicates a clinically less aggressive phenotype.
Collapse
Affiliation(s)
- Gustav J Ullenhag
- Section of Oncology, Department of Radiology, Oncology and Radiation Science, Uppsala University, Uppsala, Sweden,
| | | | | | | | | | | |
Collapse
|
46
|
Mitochondrial inhibitor sensitizes non-small-cell lung carcinoma cells to TRAIL-induced apoptosis by reactive oxygen species and Bcl-X(L)/p53-mediated amplification mechanisms. Cell Death Dis 2014; 5:e1579. [PMID: 25522273 PMCID: PMC4649849 DOI: 10.1038/cddis.2014.547] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 11/13/2014] [Accepted: 11/17/2014] [Indexed: 12/20/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising agent for anticancer therapy; however, non-small-cell lung carcinoma (NSCLC) cells are relatively TRAIL resistant. Identification of small molecules that can restore NSCLC susceptibility to TRAIL-induced apoptosis is meaningful. We found here that rotenone, as a mitochondrial respiration inhibitor, preferentially increased NSCLC cells sensitivity to TRAIL-mediated apoptosis at subtoxic concentrations, the mechanisms by which were accounted by the upregulation of death receptors and the downregulation of c-FLIP (cellular FLICE-like inhibitory protein). Further analysis revealed that death receptors expression by rotenone was regulated by p53, whereas c-FLIP downregulation was blocked by Bcl-XL overexpression. Rotenone triggered the mitochondria-derived reactive oxygen species (ROS) generation, which subsequently led to Bcl-XL downregulation and PUMA upregulation. As PUMA expression was regulated by p53, the PUMA, Bcl-XL and p53 in rotenone-treated cells form a positive feedback amplification loop to increase the apoptosis sensitivity. Mitochondria-derived ROS, however, promote the formation of this amplification loop. Collectively, we concluded that ROS generation, Bcl-XL and p53-mediated amplification mechanisms had an important role in the sensitization of NSCLC cells to TRAIL-mediated apoptosis by rotenone. The combined TRAIL and rotenone treatment may be appreciated as a useful approach for the therapy of NSCLC that warrants further investigation.
Collapse
|
47
|
Schmid D, Jarvis GE, Fay F, Small DM, Greene MK, Majkut J, Spence S, McLaughlin KM, McCloskey KD, Johnston PG, Kissenpfennig A, Longley DB, Scott CJ. Nanoencapsulation of ABT-737 and camptothecin enhances their clinical potential through synergistic antitumor effects and reduction of systemic toxicity. Cell Death Dis 2014; 5:e1454. [PMID: 25299779 PMCID: PMC4649518 DOI: 10.1038/cddis.2014.413] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 08/28/2014] [Accepted: 08/29/2014] [Indexed: 01/09/2023]
Abstract
The simultaneous delivery of multiple cancer drugs in combination therapies to achieve optimal therapeutic effects in patients can be challenging. This study investigated whether co-encapsulation of the BH3-mimetic ABT-737 and the topoisomerase I inhibitor camptothecin (CPT) in PEGylated polymeric nanoparticles (NPs) was a viable strategy for overcoming their clinical limitations and to deliver both compounds at optimal ratios. We found that thrombocytopenia induced by exposure to ABT-737 was diminished through its encapsulation in NPs. Similarly, CPT-associated leukopenia and gastrointestinal toxicity were reduced compared with the administration of free CPT. In addition to the reduction of dose-limiting side effects, the co-encapsulation of both anticancer compounds in a single NP produced synergistic induction of apoptosis in both in vitro and in vivo colorectal cancer models. This strategy may widen the therapeutic window of these and other drugs and may enhance the clinical efficacy of synergistic drug combinations.
Collapse
Affiliation(s)
- D Schmid
- School of Pharmacy, Queen's University Belfast, Belfast, UK
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - G E Jarvis
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - F Fay
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - D M Small
- Centre for Infection and Immunity, Queen's University Belfast, Belfast, UK
| | - M K Greene
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - J Majkut
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - S Spence
- Centre for Infection and Immunity, Queen's University Belfast, Belfast, UK
| | - K M McLaughlin
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - K D McCloskey
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - P G Johnston
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - A Kissenpfennig
- Centre for Infection and Immunity, Queen's University Belfast, Belfast, UK
| | - D B Longley
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - C J Scott
- School of Pharmacy, Queen's University Belfast, Belfast, UK
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| |
Collapse
|
48
|
Jang JH, Cho YC, Kim KH, Lee KS, Lee J, Kim DE, Park JS, Jang BC, Kim S, Kwon TK, Park JW. BAI, a novel Cdk inhibitor, enhances farnesyltransferase inhibitor LB42708-mediated apoptosis in renal carcinoma cells through the downregulation of Bcl-2 and c-FLIP (L). Int J Oncol 2014; 45:1680-90. [PMID: 24993441 DOI: 10.3892/ijo.2014.2534] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 05/26/2014] [Indexed: 11/06/2022] Open
Abstract
Previously, we reported the potential of a novel Cdk inhibitor, 2-[1,1'-biphenyl]-4-yl-N-[5-(1,1-dioxo-1λ6-isothiazolidin-2-yl)-1H-indazol-3-yl]acetamide (BAI) as a cancer chemotherapeutic agent. In this study, we investigated mechanisms by which BAI modulates FTI-mediated apoptosis in human renal carcinoma Caki cells. BAI synergizes with FTI to activate DEVDase, cleavage of poly ADP-ribose polymerase (PARP), and degradation of various anti-apoptotic proteins in Caki cells. BAI plus LB42708-induced apoptosis was inhibited by pretreatment with pan-caspase inhibitor, z-VAD-fmk, but not by overexpression of CrmA. The ROS scavenger, N-acetylcysteine (NAC) did not reduce BAI plus LB4270-induced apoptosis. Co-treatment of BAI and LB42708 reduced the mitochondrial membrane potential (MMP, ∆Ψm) in a time-dependent manner, and induced release of AIF and cytochrome c from mitochondria in Caki cells. Furthermore, BAL plus LB42708 induced downregulation of anti-apoptotic proteins [c-FLIP (L), c-FLIP (s), Bcl-2, XIAP, and Mcl-1 (L)]. Especially, we found that BAI plus LB42708-induced apoptosis was significantly attenuated by overexpression of Bcl-2 and partially blocked by overexpression of c-FLIP (L). Taken together, our results show that the activity of BAI plus LB42708 modulate multiple components in apoptotic response of human renal Caki cells, and indicate a potential as combinational therapeutic agents for preventing cancer such as renal carcinoma.
Collapse
Affiliation(s)
- Ji Hoon Jang
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Yoon Chul Cho
- Department of Urology, Dongguk University, College of Medicine, Gyeongju, Republic of Korea
| | - Ki Ho Kim
- Department of Urology, Dongguk University, College of Medicine, Gyeongju, Republic of Korea
| | - Kyung Seop Lee
- Department of Urology, Dongguk University, College of Medicine, Gyeongju, Republic of Korea
| | - Jinho Lee
- Department of Chemistry, Keimyung University, Daegu, Republic of Korea
| | - Dong Eun Kim
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Jun-Soo Park
- Chronic Disease Research Center, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Byeong-Churl Jang
- Chronic Disease Research Center, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Shin Kim
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Jong-Wook Park
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| |
Collapse
|
49
|
Venza I, Visalli M, Oteri R, Teti D, Venza M. Class I-specific histone deacetylase inhibitor MS-275 overrides TRAIL-resistance in melanoma cells by downregulating c-FLIP. Int Immunopharmacol 2014; 21:439-46. [PMID: 24946096 DOI: 10.1016/j.intimp.2014.05.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/06/2014] [Accepted: 05/20/2014] [Indexed: 01/03/2023]
Abstract
Tumor-necrosis factor-related apoptosis-inducing ligand (TRAIL) has selective killing effect toward malignant cells; however some human melanomas are intrinsically resistant. In this study, we have shown that class I-specific histone deacetylase inhibitor (HDACi) MS-275 can synergize with TRAIL to induce apoptosis in TRAIL-resistant cell lines and to enhance susceptibility of sensitive cells. Conversely, class II-selective HDACi MC1575 has shown no effect on the resistance of melanoma cells and was able exclusively to increase TRAIL-induced cell death in responsive cells. Both the HDACis variably increased DR4, DR5, and procaspase 8 expression, regardless whether cells were TRAIL-sensitive or TRAIL-resistant. However, only MS-275 markedly decreased the expression levels of both the long and short c-FLIP isoforms. RNAi-mediated c-FLIP silencing resulted in caspase 8-dependent apoptosis in survivor cells which was comparable to that observed following MS-275 treatment. Accordingly, enforced expression of ectopic c-FLIP has abolished the cooperative induction of apoptosis by the combination of MS-275 and TRAIL. These data indicate that c-FLIP is a critical regulator of death ligand sensitivity in melanoma. Inhibition of class I HDAC isoenzymes 1, 2 and 3 has resulted to be functionally important for c-FLIP downregulation by MS-275. In contrast, knockdown of class II HDACs has had no effect on c-FLIP expression, thus explaining the dual incapacity of MC1575 to inhibit c-FLIP expression and sensitize cells resistant to TRAIL. The data reported here suggest that MS-275 represents a promising therapeutic approach in combination with TRAIL for treatment of cutaneous and uveal melanoma due to its ability to reduce c-FLIP expression.
Collapse
Affiliation(s)
- Isabella Venza
- Department of Experimental Specialized Medical and Surgical and Odontostomatology Sciences, University of Messina, Messina, Italy
| | - Maria Visalli
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Rosaria Oteri
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Diana Teti
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.
| | - Mario Venza
- Department of Experimental Specialized Medical and Surgical and Odontostomatology Sciences, University of Messina, Messina, Italy
| |
Collapse
|
50
|
Poor antibody validation is a challenge in biomedical research: a case study for detection of c-FLIP. Apoptosis 2014; 18:1154-62. [PMID: 23917691 DOI: 10.1007/s10495-013-0880-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Successful translation of findings derived from preclinical studies into effective therapies is critical in biomedical research. Lack of robustness and reproducibility of the preclinical data, due to insufficient number of repeats, inadequate cell-based and mouse models contribute to the poor success rate. Antibodies are widely used in preclinical research, notably to determine the expression of potential therapeutic targets in tissues of interest, including tumors, but also to identify disease and/or treatment response biomarkers. We sought to determine whether the current antibody characterization standards in preclinical research are sufficient to ensure reliability of the data found in peer-reviewed publications. To address this issue, we used detection of the protein c-FLIP, a major factor of resistance to apoptosis, as a proof of concept. Accurate detection of endogenous c-FLIP levels in the preclinical settings is imperative since it is considered as a potential theranostic biomarker. Several sources of c-FLIP antibodies validated by their manufacturer and recommended for western blotting were therefore rigorously tested. We found a wide divergence in immune recognition properties. While these antibodies have been used in many publications, our results show that several of them failed to detect endogenous c-FLIP protein by Western blotting. Our results suggest that antibody validation standards are inadequate, and that systematic use of genetic knockdowns and/or knockouts to establish proof of specificity is critical, even for antibodies previously used in the scientific literature. Because antibodies are fundamental tools in both preclinical and clinical research, ensuring their specificity is crucial.
Collapse
|