1
|
Cai L, Chen H, Wang Y, Zhang J, Song D, Tan Y, Guo Z, Wang X. Platinum(IV) Complexes Trigger Death Receptors and Natural Killer Cells to Suppress Breast Cancer. J Med Chem 2025. [PMID: 39886904 DOI: 10.1021/acs.jmedchem.4c02509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Chemoimmunotherapy is an alternative treatment against cancers. Platinum(IV) complexes FMP and DFMP, coupling formononetin derivative as axial ligand(s), were designed to suppress triple-negative breast cancer (TNBC) by activating death receptors (DRs) and natural killer (NK) cells. These complexes show great potential to overcome the resistance of TNBC to chemotherapy by inducing both intrinsic and extrinsic apoptosis in cancer cells. Particularly, FMP with one axial formononetin derivative not only induced the caspase-3-dependent intrinsic apoptosis but also upregulated the expression of DRs and caspase-8, triggered the extrinsic apoptosis, and enhanced the cytotoxic ability of NK92 cells. Moreover, FMP increased the release of granzyme B, restrained the proliferation and differentiation of myeloid-derived suppressor cells, and the secretion of IL-10, thus inhibiting the TNBC in vitro and in vivo. The results demonstrate that FMP overcomes the chemoresistance and immune escape of TNBC through a new mechanism involving the synergy of chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Linxiang Cai
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Hanhua Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Ying Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Jingwen Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Dongfan Song
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yehong Tan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
2
|
Alaei A, Solali S, Mirzapour MM. Study of TRAIL and SAHA Co-Treatment on Leukemia K562 Cell Line. Cell Biochem Biophys 2024:10.1007/s12013-024-01543-y. [PMID: 39392552 DOI: 10.1007/s12013-024-01543-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 10/12/2024]
Abstract
TRAIL (Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand) is an attractive agent being considered a potential cancer treatment. It attaches to its death receptors, leading many cancer cells to apoptosis. However, some malignancies indicate substantial resistance to TRAIL, challenging anticancer scientists. Herein, combination therapy with TRAIL plus SAHA (Suberoyl Anilide Hydroxamic Acid) was conducted to evaluate the capability of SAHA to overcome TRAIL resistance in the leukemia K562 cell line. First, the IC50 for SAHA was calculated (2 µM) at 12, 24, 48, and 72 h of treatment using MTT assay. Second, the K562 cells were treated with concentrations of 50 and 100 nM of TRAIL and 2 μM of SAHA separately and together for 24, 48, and 72 h and the survival of these cells was evaluated by Flowcytometry following the annexin-V and PI staining. To demonstrate the non-toxicity of the combined treatment for normal cells, the HEK-293 cell line was treated with the TRAIL 100 nM and SAHA 2 μM combined and separated at the same periods. In the end, by performing real-time PCR, the amount of candidate genes' expression implicated in TRAIL resistance, and the levels of BCR-ABL expression was measured. The drug dosages were not toxic to normal cells. SAHA plus TRAIL strongly triggered apoptosis in K562 cells after 24, 48, and 72 h of exposure. Furthermore, it was shown that DR4, DR5, and CHOP expressions were enhanced, and PI3K, Akt, ERK, STAT3, c-FLIPL, NF-κB, and BCR-ABL expressions were decreased by SAHA in K562 cells. Our study indicated that SAHA combined with TRAIL can increase the sensitivity of K562 leukemic cells to TRAIL by suppressing intracellular anti-apoptotic molecules and augmenting the expressions of DR4/DR5 and CHOP.
Collapse
Affiliation(s)
- Amirarsalan Alaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Solali
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | |
Collapse
|
3
|
Ripoll-Viladomiu I, Prina-Mello A, Movia D, Marignol L. Extracellular vesicles and the "six Rs" in radiotherapy. Cancer Treat Rev 2024; 129:102799. [PMID: 38970839 DOI: 10.1016/j.ctrv.2024.102799] [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: 03/06/2024] [Revised: 06/14/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Over half of patients with cancer receive radiation therapy during the course of their disease. Decades of radiobiological research have identified 6 parameters affecting the biological response to radiation referred to as the 6 "Rs": Repair, Radiosensitivity, Repopulation, Redistribution, Reoxygenation, and Reactivation of the anti-tumour immune response. Extracellular Vesicles (EVs) are small membrane-bound particles whose multiple biological functions are increasingly documented. Here we discuss the evidence for a role of EVs in the orchestration of the response of cancer cells to radiotherapy. We highlight that EVs are involved in DNA repair mechanisms, modulation of cellular sensitivity to radiation, and facilitation of tumour repopulation. Moreover, EVs influence tumour reoxygenation dynamics, and play a pivotal role in fostering radioresistance. Last, we examine how EV-related strategies could be translated into novel strategies aimed at enhancing the efficacy of radiation therapy against cancer.
Collapse
Affiliation(s)
- Isabel Ripoll-Viladomiu
- Trinity St. James's Cancer Institute, Radiobiology and Molecular Oncology Research Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Ireland; Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Adriele Prina-Mello
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Dania Movia
- Trinity St. James's Cancer Institute, Radiobiology and Molecular Oncology Research Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Ireland; Department of Biology and Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Callan Building, Maynooth, Ireland
| | - Laure Marignol
- Trinity St. James's Cancer Institute, Radiobiology and Molecular Oncology Research Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Ireland.
| |
Collapse
|
4
|
Hui San S, Ching Ngai S. E-cadherin re-expression: Its potential in combating TRAIL resistance and reversing epithelial-to-mesenchymal transition. Gene 2024; 909:148293. [PMID: 38373660 DOI: 10.1016/j.gene.2024.148293] [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: 10/13/2023] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 02/21/2024]
Abstract
The major limitation of conventional chemotherapy drugs is their lack of specificity for cancer cells. As a selective apoptosis-inducing agent, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has emerged as an attractive alternative. However, most of the cancer cells are found to be either intrinsically resistant to the TRAIL protein or may develop resistance after multiple treatments, and TRAIL resistance can induce epithelial-to-mesenchymal transition (EMT) at a later stage, promoting cancer invasion and migration. Interestingly, E-cadherin loss has been linked to TRAIL resistance and initiation of EMT, making E-cadherin re-expression a potential target to overcome these obstacles. Recent research suggests that re-expressing E-cadherin may reduce TRAIL resistance by enhancing TRAIL-induced apoptosis and preventing EMT by modulating EMT signalling factors. This reversal of EMT, can also aid in improving TRAIL-induced apoptosis. Therefore, this review provides remarkable insights into the mechanisms underlying E-cadherin re-expression, clinical implications, and potentiation, as well as the research gaps of E-cadherin re-expression in the current cancer treatment.
Collapse
Affiliation(s)
- Ser Hui San
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia
| | - Siew Ching Ngai
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| |
Collapse
|
5
|
Vunnam N, Young MC, Liao EE, Lo CH, Huber E, Been M, Thomas DD, Sachs JN. Nimesulide, a COX-2 inhibitor, sensitizes pancreatic cancer cells to TRAIL-induced apoptosis by promoting DR5 clustering †. Cancer Biol Ther 2023; 24:2176692. [PMID: 36775838 PMCID: PMC9928464 DOI: 10.1080/15384047.2023.2176692] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023] Open
Abstract
Nimesulide is a nonsteroidal anti-inflammatory drug and a COX-2 inhibitor with antitumor and antiproliferative activities that induces apoptosis in oral, esophagus, breast, and pancreatic cancer cells. Despite being removed from the market due to hepatotoxicity, nimesulide is still an important research tool being used to develop new anticancer drugs. Multiple studies have been done to modify the nimesulide skeleton to develop more potent anticancer agents and related compounds are promising scaffolds for future development. As such, establishing a mechanism of action for nimesulide remains an important part of realizing its potential. Here, we show that nimesulide enhances TRAIL-induced apoptosis in resistant pancreatic cancer cells by promoting clustering of DR5 in the plasma membrane. In this way, nimesulide acts like a related compound, DuP-697, which sensitizes TRAIL-resistant colon cancer cells in a similar manner. Our approach applies a time-resolved FRET-based biosensor that monitors DR5 clustering and conformational states in the plasma membrane. We show that this tool can be used for future high-throughput screens to identify novel, nontoxic small molecule scaffolds to overcome TRAIL resistance in cancer cells.
Collapse
Affiliation(s)
- Nagamani Vunnam
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Malaney C Young
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Elly E Liao
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Chih Hung Lo
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Evan Huber
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - MaryJane Been
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Jonathan N Sachs
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
6
|
Coppola V, Marino I, Warnken U, Falchi M, Pasquini L, Biffoni M, De Maria R, Haas TL. The autophagic protein FYCO1 controls TNFRSF10/TRAIL receptor induced apoptosis and is inactivated by CASP8 (caspase 8). Autophagy 2023; 19:2733-2751. [PMID: 37418591 PMCID: PMC10472876 DOI: 10.1080/15548627.2023.2229656] [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: 05/13/2022] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023] Open
Abstract
Apoptosis is a tightly controlled cell death program executed by proteases, the so-called caspases. It plays an important role in tissue homeostasis and is often dysregulated in cancer. Here, we identified FYCO1, a protein that promotes microtubule plus end-directed transport of autophagic and endosomal vesicles as a molecular interaction partner of activated CASP8 (caspase 8). The absence of FYCO1 sensitized cells to basal and TNFSF10/TRAIL-induced apoptosis by receptor accumulation and stabilization of the Death Inducing Signaling Complex (DISC). Loss of FYCO1 resulted in impaired transport of TNFRSF10B/TRAIL-R2/DR5 (TNF receptor superfamily member 10b) to the lysosomes in TNFSF10/TRAIL-stimulated cells. More in detail, we show that FYCO1 interacted via its C-terminal GOLD domain with the CCZ1-MON1A complex, which is necessary for RAB7A activation and for the fusion of autophagosomal/endosomal vesicles with lysosomes. We demonstrated that FYCO1 is a novel and specific CASP8 substrate. The cleavage at aspartate 1306 resulted in the release of the C-terminal GOLD domain, inactivating FYCO1 function, and allowing for the progression of apoptosis. Furthermore, the lack of FYCO1 resulted in a stronger and prolonged formation of the TNFRSF1A/TNF-R1 signaling complex. Thus, FYCO1 limits the ligand-induced and steady-state signaling of TNFR-superfamily members, providing a control mechanism that fine-tunes both apoptotic and inflammatory answers.Abbreviations: AP: affinity purification; CHX: cycloheximide; co-IP: co-immunoprecipitation; CRISPR: clustered regularly interspaced short palindromic repeats; DISC: death-inducing signaling complex; DR: death receptors; doxy: doxycycline; GEF: guanine nucleotide exchange factor; ind: inducible; KD: knockdown; KO: knockout; MS: mass spectrometry; shRNA: short hairpin RNA; siRNA: small interfering RNA; TIP: two-step co-immunoprecipitation; WB: western blot.
Collapse
Affiliation(s)
- Valeria Coppola
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, RM, Italy
| | - Ilaria Marino
- Department of Translational Medicine and Surgery, Università Cattolica Del Sacro Cuore, Rome, RM, Italy
| | - Uwe Warnken
- Functional Proteomic Analysis, German Cancer Research Center (DKFZ), Heidelberg, BW, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, BW, Germany
| | | | - Luca Pasquini
- Servizio Tecnico Scientifico Grandi Strumentazioni E Core Facilities – FAST, Rome, RM, Italy
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, RM, Italy
| | - Ruggero De Maria
- Department of Translational Medicine and Surgery, Università Cattolica Del Sacro Cuore, Rome, RM, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, RM, Italy
| | - Tobias Longin Haas
- Department of Translational Medicine and Surgery, Università Cattolica Del Sacro Cuore, Rome, RM, Italy
- Section of Immunotherapy, IIGM-Italian Institute for Genomic Medicine, Candiolo, TO, Italy
| |
Collapse
|
7
|
Gunalp S, Helvaci DG, Oner A, Bursalı A, Conforte A, Güner H, Karakülah G, Szegezdi E, Sag D. TRAIL promotes the polarization of human macrophages toward a proinflammatory M1 phenotype and is associated with increased survival in cancer patients with high tumor macrophage content. Front Immunol 2023; 14:1209249. [PMID: 37809073 PMCID: PMC10551148 DOI: 10.3389/fimmu.2023.1209249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023] Open
Abstract
Background TNF-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that can either induce cell death or activate survival pathways after binding to death receptors (DRs) DR4 or DR5. TRAIL is investigated as a therapeutic agent in clinical trials due to its selective toxicity to transformed cells. Macrophages can be polarized into pro-inflammatory/tumor-fighting M1 macrophages or anti-inflammatory/tumor-supportive M2 macrophages and an imbalance between M1 and M2 macrophages can promote diseases. Therefore, identifying modulators that regulate macrophage polarization is important to design effective macrophage-targeted immunotherapies. The impact of TRAIL on macrophage polarization is not known. Methods Primary human monocyte-derived macrophages were pre-treated with either TRAIL or with DR4 or DR5-specific ligands and then polarized into M1, M2a, or M2c phenotypes in vitro. The expression of M1 and M2 markers in macrophage subtypes was analyzed by RNA sequencing, qPCR, ELISA, and flow cytometry. Furthermore, the cytotoxicity of the macrophages against U937 AML tumor targets was assessed by flow cytometry. TCGA datasets were also analyzed to correlate TRAIL with M1/M2 markers, and the overall survival of cancer patients. Results TRAIL increased the expression of M1 markers at both mRNA and protein levels while decreasing the expression of M2 markers at the mRNA level in human macrophages. TRAIL also shifted M2 macrophages towards an M1 phenotype. Our data showed that both DR4 and DR5 death receptors play a role in macrophage polarization. Furthermore, TRAIL enhanced the cytotoxicity of macrophages against the AML cancer cells in vitro. Finally, TRAIL expression was positively correlated with increased expression of M1 markers in the tumors from ovarian and sarcoma cancer patients and longer overall survival in cases with high, but not low, tumor macrophage content. Conclusions TRAIL promotes the polarization of human macrophages toward a proinflammatory M1 phenotype via both DR4 and DR5. Our study defines TRAIL as a new regulator of macrophage polarization and suggests that targeting DRs can enhance the anti-tumorigenic response of macrophages in the tumor microenvironment by increasing M1 polarization.
Collapse
Affiliation(s)
- Sinem Gunalp
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Derya Goksu Helvaci
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Aysenur Oner
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | | | - Alessandra Conforte
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Hüseyin Güner
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Science, Abdullah Gül University, Kayseri, Türkiye
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Eva Szegezdi
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Duygu Sag
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| |
Collapse
|
8
|
Tapadar P, Pal A, Ghosal N, Kumar B, Paul T, Biswas N, Pal R. CDH1 overexpression sensitizes TRAIL resistant breast cancer cells towards rhTRAIL induced apoptosis. Mol Biol Rep 2023; 50:7283-7294. [PMID: 37422537 DOI: 10.1007/s11033-023-08657-1] [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: 04/20/2023] [Accepted: 06/29/2023] [Indexed: 07/10/2023]
Abstract
PURPOSE Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is well known for its unique ability to induce apoptosis in cancer cells but not normal cells. However, a subpopulation of cancer cells exist that does not respond to toxic doses of TRAIL. In this study, we aimed to identify key factors regulating TRAIL resistance in breast cancer. METHODS rhTRAIL (recombinant human TRAIL) resistant cells (TR) isolated from TRAIL sensitive MDA-MB-231 parental cells (TS) were confirmed using trypan blue assay, cell viability assay and AO/EtBr (acridine orange/ethidium bromide) staining. Microarray was performed followed by analysis using DAVID and Cytoscape bioinformatics software to identify the candidate hub gene. Gene expression of the candidate gene was confirmed using real-time PCR and western blot. Candidate gene was overexpressed via transient transfection to identify its significance in the context of rhTRAIL. Breast cancer patient data was obtained from The Cancer Genome Atlas (TCGA) database. RESULTS Whole transcriptome analysis identified 4907 differentially expressed genes (DEGs) between TS and TR cells. CDH1 was identified as the candidate hub gene, with 18-degree centrality. We further observed CDH1 protein to be downregulated, overexpression of which increased apoptosis in TR cells after rhTRAIL treatment. TCGA patient data analysis also showed CDH1 mRNA to be low in TRAIL resistant patient group compared to TRAIL sensitive group. CONCLUSION CDH1 overexpression sensitizes TR cells towards rhTRAIL induced apoptosis. Therefore, we can hypothesize that CDH1 expression should be taken into account while performing TRAIL therapy in breast cancer.
Collapse
Affiliation(s)
- Poulami Tapadar
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700073, India
| | - Ambika Pal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700073, India
| | - Nirajan Ghosal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700073, India
| | - Bhupender Kumar
- Department of Biochemistry, Institute of Home Economics, University of Delhi, New Delhi, 110016, India
| | - Tamalika Paul
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700073, India
| | - Nabendu Biswas
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700073, India
| | - Ranjana Pal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700073, India.
| |
Collapse
|
9
|
Gampa SC, Garimella SV, Pandrangi S. Nano-TRAIL: a promising path to cancer therapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:78-102. [PMID: 37065863 PMCID: PMC10099604 DOI: 10.20517/cdr.2022.82] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/20/2022] [Accepted: 01/04/2023] [Indexed: 04/18/2023]
Abstract
Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand, also called apo-2 ligand (TRAIL/Apo-2L), is a cytokine that triggers apoptosis by binding to TRAIL-R1 (DR4) and TRAIL-R2 (DR5) death receptors. Apoptosis occurs through either the extrinsic or intrinsic pathway. The administration of recombinant human TRAIL (rhTRAIL) or TRAIL-receptor (TRAIL-R) agonists promotes apoptosis preferentially in cancerous cells over normal cells in vitro; this phenomenon has also been observed in clinical studies. The limited efficacy of rhTRAIL in clinical trials could be attributed to drug resistance, short half-life, targeted delivery issues, and off-target toxicities. Nanoparticles are excellent drug and gene delivery systems characterized by improved permeability and retention, increased stability and biocompatibility, and precision targeting. In this review, we discuss resistance mechanisms to TRAIL and methods to overcome TRAIL resistance by using nanoparticle-based formulations developed for the delivery of TRAIL peptides, TRAIL-R agonists, and TRAIL genes to cancer cells. We also discuss combinatorial approaches of chemotherapeutic drugs with TRAIL. These studies demonstrate TRAIL's potential as an anticancer agent.
Collapse
Affiliation(s)
- Siri Chandana Gampa
- Department of Biotechnology, Institute of Science, GITAM (Deemed to be University), Andhra Pradesh 530045, India
| | - Sireesha V. Garimella
- Department of Biotechnology, Institute of Science, GITAM (Deemed to be University), Andhra Pradesh 530045, India
| | - SanthiLatha Pandrangi
- Department of Biochemistry and Bioinformatics, Institute of Science, GITAM (Deemed to be University), Andhra Pradesh 530045, India
| |
Collapse
|
10
|
Charvátová S, Motais B, Czapla J, Cichoń T, Smolarczyk R, Walek Z, Giebel S, Hájek R, Bagó JR. Novel Local "Off-the-Shelf" Immunotherapy for the Treatment of Myeloma Bone Disease. Cells 2023; 12:cells12030448. [PMID: 36766789 PMCID: PMC9914109 DOI: 10.3390/cells12030448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/13/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Myeloma bone disease (MBD) is one of the major complications in multiple myeloma (MM)-the second most frequent hematologic malignancy. It is characterized by the formation of bone lesions due to the local action of proliferating MM cells, and to date, no effective therapy has been developed. In this study, we propose a novel approach for the local treatment of MBD with a combination of natural killer cells (NKs) and mesenchymal stem cells (MSCs) within a fibrin scaffold, altogether known as FINM. The unique biological properties of the NKs and MSCs, joined to the injectable biocompatible fibrin, permitted to obtain an efficient "off-the-shelf" ready-to-use composite for the local treatment of MBD. Our in vitro analyses demonstrate that NKs within FINM exert a robust anti-tumor activity against MM cell lines and primary cells, with the capacity to suppress osteoclast activity (~60%) within in vitro 3D model of MBD. Furthermore, NKs' post-thawing cytotoxic activity is significantly enhanced (~75%) in the presence of MSCs, which circumvents the decrease of NKs cytotoxicity after thawing, a well-known issue in the cryopreservation of NKs. To reduce the tumor escape, we combined FINM with other therapeutic agents (bortezomib (BZ), and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)), observing a clear therapeutic synergistic effect in vitro. Finally, the therapeutic efficacy of FINM in combination with BZ and TRAIL was assessed in a mouse model of MM, achieving 16-fold smaller tumors compared to the control group without treatment. These results suggest the potential of FINM to serve as an allogeneic "off-the-shelf" approach to improve the outcomes of patients suffering from MBD.
Collapse
Affiliation(s)
- Sandra Charvátová
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Benjamin Motais
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44102 Gliwice, Poland
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44102 Gliwice, Poland
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44102 Gliwice, Poland
| | - Zuzana Walek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Sebastian Giebel
- Department of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44102 Gliwice, Poland
| | - Roman Hájek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Juli R. Bagó
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
- Correspondence: ; Tel.: +42-(05)-97372092
| |
Collapse
|
11
|
Thang M, Mellows C, Mercer-Smith A, Nguyen P, Hingtgen S. Current approaches in enhancing TRAIL therapies in glioblastoma. Neurooncol Adv 2023; 5:vdad047. [PMID: 37215952 PMCID: PMC10195206 DOI: 10.1093/noajnl/vdad047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
Glioblastoma (GBM) is the most prevalent, aggressive, primary brain cancer in adults and continues to pose major medical challenges due in part to its high rate of recurrence. Extensive research is underway to discover new therapies that target GBM cells and prevent the inevitable recurrence in patients. The pro-apoptotic protein tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted attention as an ideal anticancer agent due to its ability to selectively kill cancer cells with minimal toxicity in normal cells. Although initial clinical evaluations of TRAIL therapies in several cancers were promising, later stages of clinical trial results indicated that TRAIL and TRAIL-based therapies failed to demonstrate robust efficacies due to poor pharmacokinetics, resulting in insufficient concentrations of TRAIL at the therapeutic site. However, recent studies have developed novel ways to prolong TRAIL bioavailability at the tumor site and efficiently deliver TRAIL and TRAIL-based therapies using cellular and nanoparticle vehicles as drug loading cargos. Additionally, novel techniques have been developed to address monotherapy resistance, including modulating biomarkers associated with TRAIL resistance in GBM cells. This review highlights the promising work to overcome the challenges of TRAIL-based therapies with the aim to facilitate improved TRAIL efficacy against GBM.
Collapse
Affiliation(s)
- Morrent Thang
- Neuroscience Center, University of North Carolina—Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina—Chapel Hill School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Clara Mellows
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina—Chapel Hill School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Alison Mercer-Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina—Chapel Hill School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Phuong Nguyen
- Michigan State University School of Medicine, East Lansing, Michigan, USA
| | - Shawn Hingtgen
- Corresponding Author: Shawn Hingtgen, PhD, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Eshelman School of Pharmacy, 125 Mason Farm Road, Chapel Hill, NC 27599-7363, USA ()
| |
Collapse
|
12
|
Bhere D, Choi SH, van de Donk P, Hope D, Gortzak K, Kunnummal A, Khalsa J, Revai Lechtich E, Reinshagen C, Leon V, Nissar N, Bi WL, Feng C, Li H, Zhang YS, Liang SH, Vasdev N, Essayed WI, Quevedo PV, Golby A, Banouni N, Palagina A, Abdi R, Fury B, Smirnakis S, Lowe A, Reeve B, Hiller A, Chiocca EA, Prestwich G, Wakimoto H, Bauer G, Shah K. Target receptor identification and subsequent treatment of resected brain tumors with encapsulated and engineered allogeneic stem cells. Nat Commun 2022; 13:2810. [PMID: 35589724 PMCID: PMC9120173 DOI: 10.1038/s41467-022-30558-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/03/2022] [Indexed: 11/09/2022] Open
Abstract
Cellular therapies offer a promising therapeutic strategy for the highly malignant brain tumor, glioblastoma (GBM). However, their clinical translation is limited by the lack of effective target identification and stringent testing in pre-clinical models that replicate standard treatment in GBM patients. In this study, we show the detection of cell surface death receptor (DR) target on CD146-enriched circulating tumor cells (CTC) captured from the blood of mice bearing GBM and patients diagnosed with GBM. Next, we developed allogeneic "off-the-shelf" clinical-grade bifunctional mesenchymal stem cells (MSCBif) expressing DR-targeted ligand and a safety kill switch. We show that biodegradable hydrogel encapsulated MSCBif (EnMSCBif) has a profound therapeutic efficacy in mice bearing patient-derived invasive, primary and recurrent GBM tumors following surgical resection. Activation of the kill switch enhances the efficacy of MSCBif and results in their elimination post-tumor treatment which can be tracked by positron emission tomography (PET) imaging. This study establishes a foundation towards a clinical trial of EnMSCBif in primary and recurrent GBM patients.
Collapse
Affiliation(s)
- Deepak Bhere
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29201, USA
| | - Sung Hugh Choi
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Pim van de Donk
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - David Hope
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kiki Gortzak
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Amina Kunnummal
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jasneet Khalsa
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Esther Revai Lechtich
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Clemens Reinshagen
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Victoria Leon
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Nabil Nissar
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Cheng Feng
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hongbin Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yu Shrike Zhang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Steven H Liang
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Neil Vasdev
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Walid Ibn Essayed
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Pablo Valdes Quevedo
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Alexandra Golby
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Naima Banouni
- Department of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Anna Palagina
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Reza Abdi
- Department of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Brian Fury
- UC Davis Institute for Regenerative Cures, Davis, CA, 95817, USA
| | - Stelios Smirnakis
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Alarice Lowe
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Brock Reeve
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Arthur Hiller
- Amasa Therapeutics Inc., 1 Harmony Lane, Andover, MA, 01810, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Glenn Prestwich
- Department of Medicinal Chemistry, College of Pharmacy University of Utah, Salt Lake City, UT, 84112, USA
- Washington State University Health Sciences, Spokane, WA, 99202, USA
| | - Hiroaki Wakimoto
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Gerhard Bauer
- UC Davis Institute for Regenerative Cures, Davis, CA, 95817, USA
| | - Khalid Shah
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
13
|
Yoshida T, Yamasaki K, Tadagaki K, Kuwahara Y, Matsumoto A, Sofovic AE, Kondo N, Sakai T, Okuda T. Tumor necrosis factor‑related apoptosis‑inducing ligand is a novel transcriptional target of runt‑related transcription factor 1. Int J Oncol 2021; 60:6. [PMID: 34958111 PMCID: PMC8727134 DOI: 10.3892/ijo.2021.5296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/23/2021] [Indexed: 11/26/2022] Open
Abstract
Runt-related transcription factor 1 (RUNX1), which is also known as acute myeloid leukemia 1 (AML1), has been frequently found with genomic aberrations in human leukemia. RUNX1 encodes a transcription factor that can regulate the expression of hematopoietic genes. In addition, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) performs an important function for malignant tumors in immune surveillance. However, the regulatory mechanism of TRAIL expression remain to be fully elucidated. In the present study, tetradecanoylphorbol 13-acetate-treated megakaryocytic differentiated K562 cells was used to examine the effect of RUNX1 on TRAIL expression. Luciferase assay series of TRAIL promoters for the cells co-transfected with RUNX1 and core-binding factor β (CBFβ) expression vectors were performed to evaluate the nature of TRAIL transcriptional regulation. Electrophoresis mobility shift assay of the RUNX1 consensus sequence of the TRAIL promoter with recombinant RUNX1 and CBFβ proteins was also performed. BloodSpot database analysis for TRAIL expression in patients with acute myeloid leukemia were performed. The expression of TRAIL, its receptor Death receptor 4 and 5 and RUNX1 in K562 cells transfected with the RUNX1 expression vector and RUNX1 siRNA were evaluated by reverse transcription-quantitative PCR (RT-qPCR). TRAIL and RUNX1-ETO expression was also measured in Kasumi-1 cells transfected with RUNX1-ETO siRNA and in KG-1 cells transfected with RUNX1-ETO expression plasmid, both by RT-qPCR. Cell counting, lactate dehydrogenase assay and cell cycle analysis by flow cytometry were performed on Kasumi-1, KG-1, SKNO-1 and K562 cells treated with TRAIL and HDAC inhibitors sodium butyrate or valproic acid. The present study demonstrated that RUNX1 is a transcriptional regulator of TRAIL. It was initially found that the induction of TRAIL expression following the megakaryocytic differentiation of human leukemia cells was RUNX1-dependent. Subsequently, overexpression of RUNX1 was found to increase TRAIL mRNA expression by activating its promoter activity. Additional analyses revealed that RUNX1 regulated the expression of TRAIL in an indirect manner, because RUNX1 retained its ability to activate this promoter following the mutation of all possible RUNX1 consensus sites. Furthermore, TRAIL expression was reduced in leukemia cells carrying the t(8;21) translocation, where the RUNX1-ETO chimeric protein interfere with normal RUNX1 function. Exogenous treatment of recombinant TRAIL proteins was found to induce leukemia cell death. To conclude, the present study provided a novel mechanism, whereby TRAIL is a target gene of RUNX1 and TRAIL expression was inhibited by RUNX1-ETO. These results suggest that TRAIL is a promising agent for the clinical treatment of t(8;21) AML.
Collapse
Affiliation(s)
- Tatsushi Yoshida
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Kenta Yamasaki
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Kenjiro Tadagaki
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Yasumichi Kuwahara
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Akifumi Matsumoto
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Adèm Ejub Sofovic
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Noriko Kondo
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Toshiyuki Sakai
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Tsukasa Okuda
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| |
Collapse
|
14
|
TRAIL Triggers CRAC-Dependent Calcium Influx and Apoptosis through the Recruitment of Autophagy Proteins to Death-Inducing Signaling Complex. Cells 2021; 11:cells11010057. [PMID: 35011619 PMCID: PMC8750441 DOI: 10.3390/cells11010057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/12/2021] [Accepted: 12/22/2021] [Indexed: 11/30/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively kills various cancer cell types, but also leads to the activation of signaling pathways that favor resistance to cell death. Here, we investigated the as yet unknown roles of calcium signaling and autophagy regulatory proteins during TRAIL-induced cell death in leukemia cells. Taking advantage of the Gene Expression Profiling Interactive Analysis (GEPIA) project, we first found that leukemia patients present a unique TRAIL receptor gene expression pattern that may reflect their resistance to TRAIL. The exposure of NB4 acute promyelocytic leukemia cells to TRAIL induces intracellular Ca2+ influx through a calcium release-activated channel (CRAC)-dependent mechanism, leading to an anti-apoptotic response. Mechanistically, we showed that upon TRAIL treatment, two autophagy proteins, ATG7 and p62/SQSTM1, are recruited to the death-inducing signaling complex (DISC) and are essential for TRAIL-induced Ca2+ influx and cell death. Importantly, the treatment of NB4 cells with all-trans retinoic acid (ATRA) led to the upregulation of p62/SQSTM1 and caspase-8 and, when added prior to TRAIL stimulation, significantly enhanced DISC formation and the apoptosis induced by TRAIL. In addition to uncovering new pleiotropic roles for autophagy proteins in controlling the calcium response and apoptosis triggered by TRAIL, our results point to novel therapeutic strategies for sensitizing leukemia cells to TRAIL.
Collapse
|
15
|
Lee YE, Yuk CM, Lee M, Han KC, Jun E, Kim TS, Ku JL, Im SG, Lee E, Jang M. Facile discovery of a therapeutic agent for NK-mediated synergistic antitumor effects using a patient-derived 3D platform. Biomater Sci 2021; 10:678-691. [PMID: 34940764 DOI: 10.1039/d1bm01699g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite the essential roles of natural killer (NK) cells in cancer treatment, the physical barrier and biological cues of the tumor microenvironment (TME) may induce NK cell dysfunction, causing their poor infiltration into tumors. The currently available two-dimensional (2D) cancer-NK co-culture systems hardly represent the characteristics of TME and are not suitable for tracking the infiltration of immune cells and assessing the efficacy of immunotherapy. This study aims to monitor NK-mediated cancer cell killing using a polymer thin film-based, 3D assay platform that contains highly tumorigenic cancer spheroids. A poly(cyclohexyl methacrylate) (pCHMA)-coated surface enables the generation of tumorigenic spheroids from pancreatic cancer patient-derived cancer cells, showing considerable amounts of extracellular matrix (ECM) proteins and cancer stem cell (CSC)-like characteristics. The 3D spheroid-based assay platform allows rapid discovery of a therapeutic agent for synergistic NK-mediated cytotoxicity through imaging-based high-content screening. In detail, the small molecule C19, known as a multi-epithelial-mesenchymal transition pathway inhibitor, is shown to enhance NK activation and infiltration via modulation of the ECM, resulting in synergistic cytotoxicity against cancer spheroids. This 3D biomimetic co-culture assay platform provides promising applications for predicting patient-specific responses to immunotherapy through advanced therapeutic combinations involving a chemical drug and immune cells.
Collapse
Affiliation(s)
- Young Eun Lee
- Theragnosis Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul 02792, Republic of Korea. .,Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Chae Min Yuk
- Theragnosis Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul 02792, Republic of Korea.
| | - Minseok Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| | - Ki-Cheol Han
- Theragnosis Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul 02792, Republic of Korea.
| | - Eunsung Jun
- Department of Convergence Medicine, Asan Institute for Life Sciences, University of Ulsan College of Medicine and Asan Medical Center, Seoul 05505, Republic of Korea.,Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Tae Sung Kim
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Ja-Lok Ku
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sung G Im
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| | - Eunjung Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| | - Mihue Jang
- Theragnosis Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul 02792, Republic of Korea. .,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
16
|
Market M, Tennakoon G, Auer RC. Postoperative Natural Killer Cell Dysfunction: The Prime Suspect in the Case of Metastasis Following Curative Cancer Surgery. Int J Mol Sci 2021; 22:ijms222111378. [PMID: 34768810 PMCID: PMC8583911 DOI: 10.3390/ijms222111378] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/02/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022] Open
Abstract
Surgical resection is the foundation for the curative treatment of solid tumors. However, metastatic recurrence due to the difficulty in eradicating micrometastases remain a feared outcome. Paradoxically, despite the beneficial effects of surgical removal of the primary tumor, the physiological stress resulting from surgical trauma serves to promote cancer recurrence and metastasis. The postoperative environment suppresses critical anti-tumor immune effector cells, including Natural Killer (NK) cells. The literature suggests that NK cells are critical mediators in the formation of metastases immediately following surgery. The following review will highlight the mechanisms that promote the formation of micrometastases by directly or indirectly inducing NK cell suppression following surgery. These include tissue hypoxia, neuroendocrine activation, hypercoagulation, the pro-inflammatory phase, and the anti-inflammatory phase. Perioperative therapeutic strategies designed to prevent or reverse NK cell dysfunction will also be examined for their potential to improve cancer outcomes by preventing surgery-induced metastases.
Collapse
Affiliation(s)
- Marisa Market
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1G 8M5, Canada; (M.M.); (G.T.)
- The Ottawa Hospital Research Institute, Ottawa, ON K1G 4E3, Canada
| | - Gayashan Tennakoon
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1G 8M5, Canada; (M.M.); (G.T.)
| | - Rebecca C. Auer
- The Ottawa Hospital Research Institute, Ottawa, ON K1G 4E3, Canada
- Department of General Surgery, The Ottawa Hospital, Ottawa, ON K1Y 4E9, Canada
- Correspondence: ; Tel.: +1-613-722-7000
| |
Collapse
|
17
|
Guesmi F, Prasad S, Ali MB, Ismail IA, Landoulsi A. Thymus hirtus sp. algeriensis Boiss. and Reut. volatile oil enhances TRAIL/Apo2L induced apoptosis and inhibits colon carcinogenesis through upregulation of death receptor pathway. Aging (Albany NY) 2021; 13:21975-21990. [PMID: 34543231 PMCID: PMC8507293 DOI: 10.18632/aging.203552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/11/2021] [Indexed: 11/25/2022]
Abstract
Background: The aim of the study is to determine the anticancer activity of Thymus algeriensis (TS) and its underlying mechanisms using in vitro and in animal models. Methods: HCT116 cells were treated with TS essential oil alone or with TRAIL, and then its anticancer effect was determined by using MTT assay, live dead assay, caspase activation and PARP cleavage. Further mechanisms of its anticancer effects was determined by analyzing expression of death receptor signaling pathway using Western blotting. A mouse model was also used to assess the antitumor potential of thyme essential oil. Results: TS oily fraction showed tumor growth inhibitory effect even at lower concentration. TS induces apoptotic cell death as indicated by cleavage of PARP, and activation of the initiator and effector caspases (caspase-3, -8 and -9). Further, results showed that TS increases the expression of death receptors (DRs) and reduces the expression of TRAIL decoy receptors (DcRs). In addition, upregulation of signaling molecules of MAPK pathway (p38 kinase, ERK, JNK), down-regulation of c-FLIP, and overexpression of SP1 and CHOP were observed by TS. Further in animal model, intragastric administration of TS (12.5 mg/ml and 50 mg/ml) prevented colorectal carcinogenesis by blocking multi-steps in carcinoma. Conclusion: Overall, these results indicate that thymus essential oil promotes apoptosis in HCT116 cells and impedes tumorigenesis in animal model. Moreover, thyme potentiates TRAIL-induced cell death through upregulation of DRs, CHOP and SP1 as well as downregulation of antiapoptotic proteins in HCT116 cells. However, therapeutic potential of TS needs to be further explored.
Collapse
Affiliation(s)
- Fatma Guesmi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Laboratory of Biochemistry and Molecular Biology, University of Carthage, Faculty of Sciences of Bizerte, Zarzouna, Bizerte 7021, Tunisia
| | - Sahdeo Prasad
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Research and Development, Noble Pharma LLC, Menomonie, WI 54751, USA
| | - Manel Ben Ali
- Department of Biology, College of Science, Taif University, Taif 21944, Saudi Arabia
| | - Ismail A Ismail
- Department of Biology, College of Science, Taif University, Taif 21944, Saudi Arabia
| | - Ahmed Landoulsi
- Laboratory of Biochemistry and Molecular Biology, University of Carthage, Faculty of Sciences of Bizerte, Zarzouna, Bizerte 7021, Tunisia
| |
Collapse
|
18
|
de Sousa E, Lérias JR, Beltran A, Paraschoudi G, Condeço C, Kamiki J, António PA, Figueiredo N, Carvalho C, Castillo-Martin M, Wang Z, Ligeiro D, Rao M, Maeurer M. Targeting Neoepitopes to Treat Solid Malignancies: Immunosurgery. Front Immunol 2021; 12:592031. [PMID: 34335558 PMCID: PMC8320363 DOI: 10.3389/fimmu.2021.592031] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 05/07/2021] [Indexed: 12/26/2022] Open
Abstract
Successful outcome of immune checkpoint blockade in patients with solid cancers is in part associated with a high tumor mutational burden (TMB) and the recognition of private neoantigens by T-cells. The quality and quantity of target recognition is determined by the repertoire of ‘neoepitope’-specific T-cell receptors (TCRs) in tumor-infiltrating lymphocytes (TIL), or peripheral T-cells. Interferon gamma (IFN-γ), produced by T-cells and other immune cells, is essential for controlling proliferation of transformed cells, induction of apoptosis and enhancing human leukocyte antigen (HLA) expression, thereby increasing immunogenicity of cancer cells. TCR αβ-dependent therapies should account for tumor heterogeneity and availability of the TCR repertoire capable of reacting to neoepitopes and functional HLA pathways. Immunogenic epitopes in the tumor-stroma may also be targeted to achieve tumor-containment by changing the immune-contexture in the tumor microenvironment (TME). Non protein-coding regions of the tumor-cell genome may also contain many aberrantly expressed, non-mutated tumor-associated antigens (TAAs) capable of eliciting productive anti-tumor immune responses. Whole-exome sequencing (WES) and/or RNA sequencing (RNA-Seq) of cancer tissue, combined with several layers of bioinformatic analysis is commonly used to predict possible neoepitopes present in clinical samples. At the ImmunoSurgery Unit of the Champalimaud Centre for the Unknown (CCU), a pipeline combining several tools is used for predicting private mutations from WES and RNA-Seq data followed by the construction of synthetic peptides tailored for immunological response assessment reflecting the patient’s tumor mutations, guided by MHC typing. Subsequent immunoassays allow the detection of differential IFN-γ production patterns associated with (intra-tumoral) spatiotemporal differences in TIL or peripheral T-cells versus TIL. These bioinformatics tools, in addition to histopathological assessment, immunological readouts from functional bioassays and deep T-cell ‘adaptome’ analyses, are expected to advance discovery and development of next-generation personalized precision medicine strategies to improve clinical outcomes in cancer in the context of i) anti-tumor vaccination strategies, ii) gauging mutation-reactive T-cell responses in biological therapies and iii) expansion of tumor-reactive T-cells for the cellular treatment of patients with cancer.
Collapse
Affiliation(s)
- Eric de Sousa
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Joana R Lérias
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Antonio Beltran
- Department of Pathology, Champalimaud Clinical Centre, Lisbon, Portugal
| | | | - Carolina Condeço
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Jéssica Kamiki
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | - Nuno Figueiredo
- Digestive Unit, Champalimaud Clinical Centre, Lisbon, Portugal
| | - Carlos Carvalho
- Digestive Unit, Champalimaud Clinical Centre, Lisbon, Portugal
| | | | - Zhe Wang
- Jiangsu Industrial Technology Research Institute (JITRI), Applied Adaptome Immunology Institute, Nanjing, China
| | - Dário Ligeiro
- Lisbon Centre for Blood and Transplantation, Instituto Português do Sangue e Transplantação (IPST), Lisbon, Portugal
| | - Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.,I Medical Clinic, Johannes Gutenberg University of Mainz, Mainz, Germany
| |
Collapse
|
19
|
Jarocka-Karpowicz I, Markowska A. Jasmonate Compounds and Their Derivatives in the Regulation of the Neoplastic Processes. Molecules 2021; 26:2901. [PMID: 34068337 PMCID: PMC8153294 DOI: 10.3390/molecules26102901] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 01/21/2023] Open
Abstract
Cancer is a serious problem in modern medicine, mainly due to the insufficient effectiveness of currently available therapies. There is a particular interest in compounds of natural origin, which can be used in the prophylaxis, as well as in the treatment and support of cancer treatment. One such compound is jasmonic acid (3-oxo-2-(pent-2'-enyl)cyclopentane acetic acid; isolated active form: trans-(-)-(3R,7R)- and cis-(+)-(3R,7S)-jasmonic acid) and its derivatives, which, due to their wide range of biological activities, are also proposed as potential therapeutic agents. Therefore, a review of literature data on the biological activity of jasmonates was prepared, with particular emphasis on the mechanisms of jasmonate action in neoplastic diseases. The anti-tumor activity of jasmonate compounds is based on altered cellular ATP levels; induction of re-differentiation through the action of Mitogen Activated Protein Kinases (MAPKs); the induction of the apoptosis by reactive oxygen species. Jasmonates can be used in anti-cancer therapy in combination with other known drugs, such as cisplatin, paclitaxel or doxorubicin, showing a synergistic effect. The structure-activity relationship of novel jasmonate derivatives with anti-tumor, anti-inflammatory and anti-aging effects is also shown.
Collapse
Affiliation(s)
- Iwona Jarocka-Karpowicz
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland;
| | | |
Collapse
|
20
|
Cytotoxic Efficacy and Resistance Mechanism of a TRAIL and VEGFA-Peptide Fusion Protein in Colorectal Cancer Models. Int J Mol Sci 2021; 22:ijms22063160. [PMID: 33808900 PMCID: PMC8003782 DOI: 10.3390/ijms22063160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 11/16/2022] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) is a type II transmembrane protein capable of selectively inducing apoptosis in cancer cells by binding to its cognate receptors. Here, we examined the anticancer efficacy of a recently developed chimeric AD-O51.4 protein, a TRAIL fused to the VEGFA-originating peptide. We tested AD-O51.4 protein activity against human colorectal cancer (CRC) models and investigated the resistance mechanism in the non-responsive CRC models. The quantitative comparison of apoptotic activity between AD-O51.4 and the native TRAIL in nine human colorectal cancer cell lines revealed dose-dependent toxicity in seven of them; the immunofluorescence-captured receptor abundance correlated with the extent of apoptosis. AD-O51.4 reduced the growth of CRC patient-derived xenografts (PDXs) with good efficacy. Cell lines that acquired AD-O51.4 resistance showed a significant decrease in surface TRAIL receptor expression and apoptosis-related proteins, including Caspase-8, HSP60, and p53. These results demonstrate the effectiveness of AD-O51.4 protein in CRC preclinical models and identify the potential mechanism underlying acquired resistance. Progression of AD-O51.4 to clinical trials is expected.
Collapse
|
21
|
Samra YA, Amin MN, Said E. Cardio-protective impact of gabapentin against doxorubicin-induced myocardial toxicity in rats; emphasis on modulation of inflammatory-apoptotic signaling. Int Immunopharmacol 2021; 90:107125. [PMID: 33199237 DOI: 10.1016/j.intimp.2020.107125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/10/2020] [Accepted: 10/15/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Cardiotoxicity is one of the most commonly encountered adverse effects observed alongside the therapeutic use of doxorubicin (DOX), thus curbing its therapeutic utility. METHODS The current study was conducted to evaluate the cardioprotective effect of gabapentin (Gaba), a Ca + 2 channel blocker with emerging pharmacological merits, against DOX-induced cardiotoxicity. Gaba was orally administered at two dose levels (10 and 30 mg/kg) for 21 days parallel to DOX injection. RESULTS DOX induced significant functional, biochemical, and histopathological injury to the myocardium. Gaba treatment revealed a cardioprotective effect as manifested in the significant restoration of electrocardiogram parameters, including the heart rate, ST segment elevation, QRS and T wave amplitudes, and QT and PR intervals. The biomarkers of myocardial injury, namely serum creatine kinase, aspartate aminotransferase, and lactate dehydrogenase activities, significantly declined as well as the concomitant improvement of the myocardial oxidative status. Mechanistically, Gaba treatment significantly reduced the myocardial contents of c-Jun N-terminal kinase (JNK), the major modulator of inflammatory/apoptotic signaling. However, the myocardial contents of the apoptotic biomarkers caspase-8 and TRAIL also significantly declined. In isolated cardiomyocytes, Gaba treatment maintained the morphological characteristics of the cardiomyocytes and preserved their spontaneous beating characteristics. Nevertheless, the protein expression of caspase-8, JNK 1/2, and CD95L significantly declined with Gaba treatment. CONCLUSION Gaba confers cardioprotective effects against DOX-induced myocardial injury and cardiotoxicity by modulating the inflammatory/apoptotic signaling pathway.
Collapse
Affiliation(s)
- Yara A Samra
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura University, 35516 Mansoura, Egypt
| | - Mohamed N Amin
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura University, 35516 Mansoura, Egypt
| | - Eman Said
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt.
| |
Collapse
|
22
|
Transferrin-Bound Doxorubicin Enhances Apoptosis and DNA Damage through the Generation of Pro-Inflammatory Responses in Human Leukemia Cells. Int J Mol Sci 2020; 21:ijms21249390. [PMID: 33321722 PMCID: PMC7764702 DOI: 10.3390/ijms21249390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022] Open
Abstract
Doxorubicin (DOX) is an effective antineoplastic drug against many solid tumors and hematological malignancies. However, the clinical use of DOX is limited, because of its unspecific mode of action. Since leukemia cells overexpress transferrin (Tf) receptors on their surface, we proposed doxorubicin–transferrin (DOX–Tf) conjugate as a new vehicle to increase drug concentration directly in cancer cells. The data obtained after experiments performed on K562 and CCRF-CEM human leukemia cell lines clearly indicate severe cytotoxic and genotoxic properties of the conjugate drug. On the other hand, normal peripheral blood mononuclear cells (PBMCs) were more resistant to DOX–Tf than to DOX. In comparison to free drug, we observed that Tf-bound DOX induced apoptosis in a TRAIL-dependent manner and caused DNA damage typical of programmed cell death. These fatal hallmarks of cell death were confirmed upon morphological observation of cells incubated with DOX or DOX–Tf. Studies of expression of TNF-α, IL-4, and IL-6 at the mRNA and protein levels revealed that the pro-inflammatory response plays an important role in the toxicity of the conjugate. Altogether, the results demonstrated here describe a mechanism of the antitumor activity of the DOX–Tf conjugate.
Collapse
|
23
|
Deng D, Shah K. TRAIL of Hope Meeting Resistance in Cancer. Trends Cancer 2020; 6:989-1001. [PMID: 32718904 PMCID: PMC7688478 DOI: 10.1016/j.trecan.2020.06.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/28/2020] [Accepted: 06/30/2020] [Indexed: 02/08/2023]
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) induces apoptosis selectively via its interaction with the death receptors TRAILR1/DR4 and TRAILR2/DR5 in a wide range of cancers, while sparing normal cells. Despite its tremendous potential for cancer therapeutics, the translation of TRAIL into the clinic has been confounded by TRAIL-resistant cancer populations. We discuss different molecular mechanisms underlying TRAIL-mediated apoptosis and resistance to TRAIL. We also discuss the successes and failures of recent preclinical and clinical studies of TRAIL-induced apoptosis, and current attempts to overcome TRAIL resistance, and we provide a perspective for improving the prospects of future clinical implementation.
Collapse
Affiliation(s)
- David Deng
- Center for Stem Cell Therapeutics and Imaging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02129, USA; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02129, USA; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02129, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
24
|
Liu Z, Li S, Ma T, Zeng J, Zhou X, Li H, Tang M, Liu X, Li F, Jiang B, Zhao M, Chen Y. Secreted TRAIL gene-modified adipose-derived stem cells exhibited potent tumor-suppressive effect in hepatocellular carcinoma cells. IMMUNITY INFLAMMATION AND DISEASE 2020; 9:144-156. [PMID: 33156578 PMCID: PMC7860607 DOI: 10.1002/iid3.372] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/20/2020] [Indexed: 01/03/2023]
Abstract
Objective Considering the potential of adipose‐derived stem cells (ADSCs) migrating towards cancer cells, this study was performed to explore the function of tumor necrosis factor‐related apoptosis‐inducing ligand (TRAIL) modified ADSCs on the development and progression of hepatocellular carcinoma (HCC). Methods ADSCs were extracted from human adipose tissues and identified through immunofluorescence and flow cytometry. Oil red staining and alizarin red staining were performed to clarify the differentiation potential of ADSCs. AAV‐CMV‐sTRAIL was transfected into ADSCs before Western blot and Transwell measurements. sTRAIL‐ADSCs were cocultured with HCC cells to explore its effect on the proliferation and apoptosis of HCC cells. The possible effect of sTRAIL‐ADSCs or ADSCs on tumor growth and metastasis was determined in vivo using xenograft nude mouse models. Results ADSCs were successfully extracted from adipose tissues, which were confirmed by cell morphology and positive expressions of CD44 and CD105. ADSCs were found with differentiation potential. After transfection, TRAIL was stably expressed in sTRAIL‐ADSCs. Both ADSCs and sTRAIL‐ADSCs can migrate towards HCC cells. In addition, sTRAIL‐ADSCs can promote the cell apoptosis and inhibit cell proliferation in vitro, on parallel it can also suppress epithelial‐mesenchymal transition, tumor growth, and metastasis in vivo. Conclusion TRAIL modified ADSCs can migrate towards HCC cells to inhibit tumor growth and the metastasis of implanted HCC tumors, which hints TRAIL modified ADSCs may be a new therapeutic approach for HCC treatment.
Collapse
Affiliation(s)
- Zhuo Liu
- Depatment of General Surgery (Three), Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Shaojie Li
- Depatment of General Surgery (Three), Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Tiexiang Ma
- Depatment of General Surgery (Three), Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Jian Zeng
- Depatment of General Surgery (Three), Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Xin Zhou
- Depatment of General Surgery (Three), Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Huanyu Li
- Depatment of General Surgery (Three), Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Min Tang
- Department of Oncology (One), Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Xiang Liu
- Depatment of General Surgery (Three), Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Feng Li
- Depatment of General Surgery (Three), Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Bin Jiang
- Depatment of General Surgery (Three), Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Ming Zhao
- Depatment of General Surgery (Three), Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Ying Chen
- Depatment of General Surgery (Three), Xiangtan Central Hospital, Xiangtan, Hunan, China
| |
Collapse
|
25
|
Engler MJ, Mimura J, Yamazaki S, Itoh K. JDP2 is directly regulated by ATF4 and modulates TRAIL sensitivity by suppressing the ATF4-DR5 axis. FEBS Open Bio 2020; 10:2771-2779. [PMID: 33108704 PMCID: PMC7714084 DOI: 10.1002/2211-5463.13017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
Jun dimerization protein 2 (JDP2) is a bZip‐type transcription factor, which acts as a repressor or activator of several cellular processes, including cell differentiation and chromatin remodeling. Previously, we found that a stress‐responsive transcription factor, known as activating transcription factor 4 (ATF4), enhances JDP2 gene expression in human astrocytoma U373MG and cervical cancer HeLa cells; however, the role of JDP2 in the ATF4‐mediated stress response remained unclear. Here, we reported that siRNA‐mediated JDP2 knockdown enhances the expression of several ATF4 target genes, including ASNS, and death receptors 4 and 5 (DR4 and DR5) in HeLa cells. In addition, the results of a transient reporter assay indicate that JDP2 overexpression represses ER stress‐mediated DR5 promoter activation suggesting that JDP2 negatively regulates ATF4‐mediated gene expression. Curiously, knockdown of JDP2 increases the sensitivity of cells to TNF‐related apoptosis‐inducing ligand (TRAIL), which induces apoptosis in cancer cells through DR4 and DR5. These results indicate that JDP2 functions as a negative feedback regulator of the ATF4 pathway and contributes to TRAIL resistance in cancer cells.
Collapse
Affiliation(s)
- Máté János Engler
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, Japan
| | - Junsei Mimura
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, Japan
| | - Shun Yamazaki
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, Japan
| | - Ken Itoh
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, Japan
| |
Collapse
|
26
|
Sciarrillo R, Wojtuszkiewicz A, Assaraf YG, Jansen G, Kaspers GJL, Giovannetti E, Cloos J. The role of alternative splicing in cancer: From oncogenesis to drug resistance. Drug Resist Updat 2020; 53:100728. [PMID: 33070093 DOI: 10.1016/j.drup.2020.100728] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022]
Abstract
Alternative splicing is a tightly regulated process whereby non-coding sequences of pre-mRNA are removed and protein-coding segments are assembled in diverse combinations, ultimately giving rise to proteins with distinct or even opposing functions. In the past decade, whole genome/transcriptome sequencing studies revealed the high complexity of splicing regulation, which occurs co-transcriptionally and is influenced by chromatin status and mRNA modifications. Consequently, splicing profiles of both healthy and malignant cells display high diversity and alternative splicing was shown to be widely deregulated in multiple cancer types. In particular, mutations in pre-mRNA regulatory sequences, splicing regulators and chromatin modifiers, as well as differential expression of splicing factors are important contributors to cancer pathogenesis. It has become clear that these aberrations contribute to many facets of cancer, including oncogenic transformation, cancer progression, response to anticancer drug treatment as well as resistance to therapy. In this respect, alternative splicing was shown to perturb the expression a broad spectrum of relevant genes involved in drug uptake/metabolism (i.e. SLC29A1, dCK, FPGS, and TP), activation of nuclear receptor pathways (i.e. GR, AR), regulation of apoptosis (i.e. MCL1, BCL-X, and FAS) and modulation of response to immunotherapy (CD19). Furthermore, aberrant splicing constitutes an important source of novel cancer biomarkers and the spliceosome machinery represents an attractive target for a novel and rapidly expanding class of therapeutic agents. Small molecule inhibitors targeting SF3B1 or splice factor kinases were highly cytotoxic against a wide range of cancer models, including drug-resistant cells. Importantly, these effects are enhanced in specific cancer subsets, such as splicing factor-mutated and c-MYC-driven tumors. Furthermore, pre-clinical studies report synergistic effects of spliceosome modulators in combination with conventional antitumor agents. These strategies based on the use of low dose splicing modulators could shift the therapeutic window towards decreased toxicity in healthy tissues. Here we provide an extensive overview of the latest findings in the field of regulation of splicing in cancer, including molecular mechanisms by which cancer cells harness alternative splicing to drive oncogenesis and evade anticancer drug treatment as well as splicing-based vulnerabilities that can provide novel treatment opportunities. Furthermore, we discuss current challenges arising from genome-wide detection and prediction methods of aberrant splicing, as well as unravelling functional relevance of the plethora of cancer-related splicing alterations.
Collapse
Affiliation(s)
- Rocco Sciarrillo
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands; Department of Pediatric Oncology, Emma's Children's Hospital, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands; Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Anna Wojtuszkiewicz
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Gerrit Jansen
- Amsterdam Immunology and Rheumatology Center, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Gertjan J L Kaspers
- Department of Pediatric Oncology, Emma's Children's Hospital, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands; Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands; Fondazione Pisana per la Scienza, Pisa, Italy
| | - Jacqueline Cloos
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
27
|
Lee WH, Han MW, Kim SH, Seong D, An JH, Chang HW, Kim SY, Kim SW, Lee JC. Tristetraprolin Posttranscriptionally Downregulates TRAIL Death Receptors. Cells 2020; 9:cells9081851. [PMID: 32784606 PMCID: PMC7465502 DOI: 10.3390/cells9081851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 12/30/2022] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has attracted attention as a potential candidate for cancer therapy. However, many primary cancers are resistant to TRAIL, even when combined with standard chemotherapy. The mechanism of TRAIL resistance in cancer cells has not been fully elucidated. The TRAIL death receptor (DR) 3′-untranslated region (3′-UTR) is reported to contain AU-rich elements (AREs) that are important for regulating DR mRNA stability. However, the mechanisms by which DR mRNA stability is determined by its 3′-UTR are unknown. We demonstrate that tristetraprolin (TTP), an ARE-binding protein, has a critical function of regulating DR mRNA stability. DR4 mRNA contains three AREs and DR5 mRNA contains four AREs in 3′-UTR. TTP bound to all three AREs in DR4 and ARE3 in DR5 and enhanced decay of DR4/5 mRNA. TTP overexpression in colon cancer cells changed the TRAIL-sensitive cancer cells to TRAIL-resistant cells, and down-regulation of TTP increased TRAIL sensitivity via DR4/5 expression. Therefore, this study provides a molecular mechanism for enhanced levels of TRAIL DRs in cancer cells and a biological basis for posttranscriptional modification of TRAIL DRs. In addition, TTP status might be a biomarker for predicting TRAIL response when a TRAIL-based treatment is used for cancer.
Collapse
Affiliation(s)
- Won Hyeok Lee
- Department of Otorhinolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea; (W.H.L.); (M.W.H.); (S.H.K.); (D.S.); (J.H.A.)
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea
| | - Myung Woul Han
- Department of Otorhinolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea; (W.H.L.); (M.W.H.); (S.H.K.); (D.S.); (J.H.A.)
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea
| | - Song Hee Kim
- Department of Otorhinolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea; (W.H.L.); (M.W.H.); (S.H.K.); (D.S.); (J.H.A.)
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea
| | - Daseul Seong
- Department of Otorhinolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea; (W.H.L.); (M.W.H.); (S.H.K.); (D.S.); (J.H.A.)
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea
| | - Jae Hee An
- Department of Otorhinolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea; (W.H.L.); (M.W.H.); (S.H.K.); (D.S.); (J.H.A.)
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea
| | - Hyo Won Chang
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.W.C.); (S.Y.K.)
| | - Sang Yoon Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.W.C.); (S.Y.K.)
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Correspondence: (S.W.K.); (J.C.L.); Tel.: +82-2-3010-4270 (S.W.K.); +82-33-610-4372 (J.C.L.); Fax: +82-2-3010-4249 (S.W.K.); +82-33-642-3189 (J.C.L.)
| | - Jong Cheol Lee
- Department of Otorhinolaryngology, GangNeung Asan Hospital, University of Ulsan College of Medicine, Gangneung 25440, Korea
- Correspondence: (S.W.K.); (J.C.L.); Tel.: +82-2-3010-4270 (S.W.K.); +82-33-610-4372 (J.C.L.); Fax: +82-2-3010-4249 (S.W.K.); +82-33-642-3189 (J.C.L.)
| |
Collapse
|
28
|
Cao Y, Kong S, Xin Y, Meng Y, Shang S, Qi Y. Lestaurtinib potentiates TRAIL-induced apoptosis in glioma via CHOP-dependent DR5 induction. J Cell Mol Med 2020; 24:7829-7840. [PMID: 32441887 PMCID: PMC7348155 DOI: 10.1111/jcmm.15415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/19/2020] [Accepted: 05/03/2020] [Indexed: 12/22/2022] Open
Abstract
Lestaurtinib, also called CEP-701, is an inhibitor of tyrosine kinase, causes haematological remission in patients with AML possessing FLT3-ITD (FLT3 gene) internal tandem duplication and strongly inhibits tyrosine kinase FLT3. Treatment with lestaurtinib modulates various signalling pathways and leads to cell growth arrest and programmed cell death in several tumour types. However, the effect of lestaurtinib on glioma remains unclear. In this study, we examined lestaurtinib and TRAIL interactions in glioma cells and observed their synergistic activity on glioma cell apoptosis. While U87 and U251 cells showed resistance to TRAIL single treatment, they were sensitized to apoptosis induced by TRAIL in the presence of lestaurtinib because of increased death receptor 5 (DR5) levels through CHOP-dependent manner. We also demonstrated using a xenograft model of mouse that the tumour growth was absolutely suppressed because of the combined treatment compared to TRAIL or lestaurtinib treatment carried out singly. Our findings reveal a potential new strategy to improve antitumour activity induced by TRAIL in glioma cells using lestaurtinib through a mechanism dependent on CHOP.
Collapse
Affiliation(s)
- Yingxiao Cao
- Department of NeurosurgeryXingtai People’s HospitalXingtaiChina
| | - Shiqi Kong
- Department of NeurosurgeryXingtai People’s HospitalXingtaiChina
| | - Yuling Xin
- Department of NeurosurgeryXingtai People’s HospitalXingtaiChina
| | - Yan Meng
- Department of Operating RoomXingtai People’s HospitalXingtaiChina
| | - Shuling Shang
- Department of Operating RoomXingtai People’s HospitalXingtaiChina
| | - Yanhui Qi
- Department of Intensive Care UnitXingtai People’s HospitalXingtaiChina
| |
Collapse
|
29
|
Liu L, Wang Q, Qiu Z, Kang Y, Liu J, Ning S, Yin Y, Pang D, Xu S. Noncoding RNAs: the shot callers in tumor immune escape. Signal Transduct Target Ther 2020; 5:102. [PMID: 32561709 PMCID: PMC7305134 DOI: 10.1038/s41392-020-0194-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 01/17/2023] Open
Abstract
Immunotherapy, designed to exploit the functions of the host immune system against tumors, has shown considerable potential against several malignancies. However, the utility of immunotherapy is heavily limited due to the low response rate and various side effects in the clinical setting. Immune escape of tumor cells may be a critical reason for such low response rates. Noncoding RNAs (ncRNAs) have been identified as key regulatory factors in tumors and the immune system. Consequently, ncRNAs show promise as targets to improve the efficacy of immunotherapy in tumors. However, the relationship between ncRNAs and tumor immune escape (TIE) has not yet been comprehensively summarized. In this review, we provide a detailed account of the current knowledge on ncRNAs associated with TIE and their potential roles in tumor growth and survival mechanisms. This review bridges the gap between ncRNAs and TIE and broadens our understanding of their relationship, providing new insights and strategies to improve immunotherapy response rates by specifically targeting the ncRNAs involved in TIE.
Collapse
Affiliation(s)
- Lei Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Qin Wang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhilin Qiu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yujuan Kang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Jiena Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Shipeng Ning
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yanling Yin
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China. .,Heilongjiang Academy of Medical Sciences, Harbin, 150086, China.
| | - Shouping Xu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
30
|
Hoshika S, Sun X, Kuranaga E, Umetsu D. Reduction of endocytic activity accelerates cell elimination during tissue remodeling of the Drosophila epidermal epithelium. Development 2020; 147:dev.179648. [PMID: 32156754 DOI: 10.1242/dev.179648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 02/24/2020] [Indexed: 12/22/2022]
Abstract
Epithelial tissues undergo cell turnover both during development and for homeostatic maintenance. Cells that are no longer needed are quickly removed without compromising the barrier function of the tissue. During metamorphosis, insects undergo developmentally programmed tissue remodeling. However, the mechanisms that regulate this rapid tissue remodeling are not precisely understood. Here, we show that the temporal dynamics of endocytosis modulate physiological cell properties to prime larval epidermal cells for cell elimination. Endocytic activity gradually reduces as tissue remodeling progresses. This reduced endocytic activity accelerates cell elimination through the regulation of Myosin II subcellular reorganization, junctional E-cadherin levels, and caspase activation. Whereas the increased Myosin II dynamics accelerates cell elimination, E-cadherin plays a protective role against cell elimination. Reduced E-cadherin is involved in the amplification of caspase activation by forming a positive-feedback loop with caspase. These findings reveal the role of endocytosis in preventing cell elimination and in the cell-property switching initiated by the temporal dynamics of endocytic activity to achieve rapid cell elimination during tissue remodeling.
Collapse
Affiliation(s)
- Shinichiro Hoshika
- Laboratory for Histogenetic Dynamics, Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Xiaofei Sun
- Laboratory for Histogenetic Dynamics, Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Erina Kuranaga
- Laboratory for Histogenetic Dynamics, Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Daiki Umetsu
- Laboratory for Histogenetic Dynamics, Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| |
Collapse
|
31
|
Kwon OS, Jung JH, Shin EA, Park JE, Park WY, Kim SH. Epigallocatechin-3-Gallate Induces Apoptosis as a TRAIL Sensitizer via Activation of Caspase 8 and Death Receptor 5 in Human Colon Cancer Cells. Biomedicines 2020; 8:biomedicines8040084. [PMID: 32283836 PMCID: PMC7235876 DOI: 10.3390/biomedicines8040084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 01/20/2023] Open
Abstract
Though epigallocatechin-3-gallate (EGCG), a major compound of green tea, has anti-diabetes, anti-obesity, anti-inflammatory, and antitumor effects, the underlying antitumor molecular mechanism of EGCG was not fully understood so far. Here the sensitizing effect of EGCG to tumor-necrosis-factor-related apoptosis-inducing ligand (TRAIL) was examined in colorectal cancers. Cotreatment of EGCG and TRAIL synergistically enhanced cytotoxicity and sub G1 accumulation, increased the number of terminal deoxynucleotidyl transferase-dT-mediated dUTP nick end labelling (TUNEL)-positive cells in SW480 and HCT116 cells. Furthermore, this cotreatment promoted the cleavages of poly (adenosine diphosphate-ribose) polymerase (PARP) and induced caspase 8 activation compared to TRAIL or EGCG alone in SW480 and HCT116 cells. Of note, cotreatment of EGCG and TRAIL increased the expression of death receptor 5 (DR5) at protein and mRNA levels and also DR5 cell surface level in colon cancer cells. Conversely, depletion of DR5 reduced the apoptotic activity of cotreatment of EGCG and TRAIL to increase cytotoxicity, sub-G1 population and PARP cleavages in colon cancer cells. Overall, our findings provide evidence that EGCG can be a sensitizer of TRAIL via DR5 and caspase 8 mediated apoptosis in colorectal cancer cells.
Collapse
|
32
|
Abstract
Calreticulin (CRT) is a pleiotropic and highly conserved molecule that is mainly localized in the endoplasmic reticulum. Recently, CRT has gained special interest for its functions outside the endoplasmic reticulum where it has immunomodulatory properties. CRT translocation to the cell membrane serves as an "eat me" signal and promotes efferocytosis of apoptotic cells and cancer cell removal with completely opposite outcomes. Efferocytosis results in a silenced immune response and homeostasis, while removal of dying cancer cells brought about by anthracycline treatment, ionizing-irradiation or photodynamic therapy results in immunogenic cell death with activation of the innate and adaptive immune responses. In addition, CRT impacts phagocyte activation and cytokine production. The effects of CRT on cytokine production depend on its conformation, species specificity, degree of oligomerization and/or glycosylation, as well as its cellular localization and the molecular partners involved. The controversial roles of CRT in cancer progression and the possible role of the CALR gene mutations in myeloproliferative neoplasms are also addressed. The release of CRT and its influence on the different cells involved during efferocytosis and immunogenic cell death points to additional roles of CRT besides merely acting as an "eat me" signal during apoptosis. Understanding the contribution of CRT in physiological and pathological processes could give us some insight into the potential of CRT as a therapeutic target.
Collapse
|
33
|
Kojima Y, Nishina T, Nakano H, Okumura K, Takeda K. Inhibition of Importin β1 Augments the Anticancer Effect of Agonistic Anti-Death Receptor 5 Antibody in TRAIL-resistant Tumor Cells. Mol Cancer Ther 2020; 19:1123-1133. [PMID: 32156787 DOI: 10.1158/1535-7163.mct-19-0597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 12/02/2019] [Accepted: 03/04/2020] [Indexed: 11/16/2022]
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) and an agonistic antibody against the death-inducing TRAIL receptor 5, DR5, are thought to selectively induce tumor cell death and therefore, have gained attention as potential therapeutics currently under investigation in several clinical trials. However, some tumor cells are resistant to TRAIL/DR5-induced cell death, even though they express DR5. Previously, we reported that DR5 is transported into the nucleus by importin β1, and knockdown of importin β1 upregulates cell surface expression of DR5 resulting in increased TRAIL sensitivity in vitro Here, we examined the impact of importin β1 knockdown on agonistic anti-human DR5 (hDR5) antibody therapy. Drug-inducible importin β1 knockdown sensitizes HeLa cells to TRAIL-induced cell death in vitro, and exerts an antitumor effect when combined with agonistic anti-hDR5 antibody administration in vivo Therapeutic importin β1 knockdown, administered via the atelocollagen delivery system, as well as treatment with the importin β inhibitor, importazole, induced regression and/or eradication of two human TRAIL-resistant tumor cells when combined with agonistic anti-hDR5 antibody treatment. Thus, these findings suggest that the inhibition of importin β1 would be useful to improve the therapeutic effects of agonistic anti-hDR5 antibody against TRAIL-resistant cancers.
Collapse
Affiliation(s)
- Yuko Kojima
- Laboratory of Morphology and Image Analysis, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Takashi Nishina
- Department of Biochemistry, Toho University School of Medicine, Tokyo, Japan
| | - Hiroyasu Nakano
- Department of Biochemistry, Toho University School of Medicine, Tokyo, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Biofunctional Microbiota, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuyoshi Takeda
- Department of Biofunctional Microbiota, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Laboratory of Cell Biology, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
34
|
Khoury L, Zalko D, Audebert M. Evaluation of the genotoxic potential of apoptosis inducers with the γH2AX assay in human cells. Mutat Res 2020; 852:503165. [PMID: 32265046 DOI: 10.1016/j.mrgentox.2020.503165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/26/2020] [Accepted: 02/28/2020] [Indexed: 11/19/2022]
Abstract
Human risk assessment of genotoxic chemicals is an important area of research. However, the specificity of in vitro mammalian genotoxicity assays is sometime low, as they yield to misleading positive results that are not observe in in vivo studies. Apoptosis can be a confounding factor in the interpretation of the results. Recently, a new strategy for genotoxicity screening, based on the combined analysis of phosphorylated histones H2AX (γH2AX) and H3 (pH3), was proposed to discriminate efficiently aneugenic from clastogenic compounds. However, γH2AX biomarker could also be induce by apoptosis. The aim of the present study was to investigate the specificity of this genotoxic biomarker. For this purpose, we analyzed 26 compounds inducing apoptosis by different mechanism of action, with the γH2AX assay in three human cell lines after 24 h treatment. Most of the tested chemicals were negative in the assay, whatever the cell line tested. The few compounds that generated positive data have also been report positive in other genotoxicity assays. The data presented here demonstrate that the γH2AX assay is not vulnerable to the generation of misleading positive results by apoptosis inducers. Currently, no formal guidelines have been approve for the γH2AX assay for regular genotoxicity studies, but we suggest that this biomarker could be used as a new standard genotoxicity assay.
Collapse
Affiliation(s)
- Laure Khoury
- Toxalim, Université de Toulouse, INRAE, INP-ENVT, INP-EI-Purpan, Université de Toulouse 3 Paul Sabatier, Toulouse, France
| | - Daniel Zalko
- Toxalim, Université de Toulouse, INRAE, INP-ENVT, INP-EI-Purpan, Université de Toulouse 3 Paul Sabatier, Toulouse, France
| | - Marc Audebert
- Toxalim, Université de Toulouse, INRAE, INP-ENVT, INP-EI-Purpan, Université de Toulouse 3 Paul Sabatier, Toulouse, France.
| |
Collapse
|
35
|
Baskar R, Fienberg HG, Khair Z, Favaro P, Kimmey S, Green DR, Nolan GP, Plevritis S, Bendall SC. TRAIL-induced variation of cell signaling states provides nonheritable resistance to apoptosis. Life Sci Alliance 2019; 2:e201900554. [PMID: 31704709 PMCID: PMC6848270 DOI: 10.26508/lsa.201900554] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 02/06/2023] Open
Abstract
TNFα-related apoptosis-inducing ligand (TRAIL), specifically initiates programmed cell death, but often fails to eradicate all cells, making it an ineffective therapy for cancer. This fractional killing is linked to cellular variation that bulk assays cannot capture. Here, we quantify the diversity in cellular signaling responses to TRAIL, linking it to apoptotic frequency across numerous cell systems with single-cell mass cytometry (CyTOF). Although all cells respond to TRAIL, a variable fraction persists without apoptotic progression. This cell-specific behavior is nonheritable where both the TRAIL-induced signaling responses and frequency of apoptotic resistance remain unaffected by prior exposure. The diversity of signaling states upon exposure is correlated to TRAIL resistance. Concomitantly, constricting the variation in signaling response with kinase inhibitors proportionally decreases TRAIL resistance. Simultaneously, TRAIL-induced de novo translation in resistant cells, when blocked by cycloheximide, abrogated all TRAIL resistance. This work highlights how cell signaling diversity, and subsequent translation response, relates to nonheritable fractional escape from TRAIL-induced apoptosis. This refined view of TRAIL resistance provides new avenues to study death ligands in general.
Collapse
Affiliation(s)
- Reema Baskar
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Harris G Fienberg
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Zumana Khair
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Patricia Favaro
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sam Kimmey
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Developmental Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Garry P Nolan
- Baxter Laboratory, Stanford University School of Medicine, Stanford, CA, USA
| | - Sylvia Plevritis
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Sean C Bendall
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
36
|
Wong SHM, Kong WY, Fang CM, Loh HS, Chuah LH, Abdullah S, Ngai SC. The TRAIL to cancer therapy: Hindrances and potential solutions. Crit Rev Oncol Hematol 2019; 143:81-94. [PMID: 31561055 DOI: 10.1016/j.critrevonc.2019.08.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/15/2022] Open
Abstract
Apoptosis is an ordered and orchestrated cellular process that occurs in physiological and pathological conditions. Resistance to apoptosis is a hallmark of virtually all malignancies. Despite being a cause of pathological conditions, apoptosis could be a promising target in cancer treatment. Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), also known as Apo-2 ligand (Apo2L), is a member of TNF cytokine superfamily. It is a potent anti-cancer agent owing to its specific targeting towards cancerous cells, while sparing normal cells, to induce apoptosis. However, resistance occurs either intrinsically or after multiple treatments which may explain why cancer therapy fails. This review summarizes the apoptotic mechanisms via extrinsic and intrinsic apoptotic pathways, as well as the apoptotic resistance mechanisms. It also reviews the current clinically tested recombinant human TRAIL (rhTRAIL) and TRAIL receptor agonists (TRAs) against TRAIL-Receptors, TRAIL-R1 and TRAIL-R2, in which the outcomes of the clinical trials have not been satisfactory. Finally, this review discusses the current strategies in overcoming resistance to TRAIL-induced apoptosis in pre-clinical and clinical settings.
Collapse
Affiliation(s)
- Sonia How Ming Wong
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Wei Yang Kong
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Hwei-San Loh
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Lay-Hong Chuah
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia; Advanced Engineering Platform, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Syahril Abdullah
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, 43400 UPM, Malaysia; UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor, 43400 UPM, Malaysia
| | - Siew Ching Ngai
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia.
| |
Collapse
|
37
|
Kong WY, Yee ZY, Mai CW, Fang CM, Abdullah S, Ngai SC. Zebularine and trichostatin A sensitized human breast adenocarcinoma cells towards tumor necrosis factor-related apoptosis inducing ligand (TRAIL)-induced apoptosis. Heliyon 2019; 5:e02468. [PMID: 31687564 PMCID: PMC6819948 DOI: 10.1016/j.heliyon.2019.e02468] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/12/2019] [Accepted: 09/09/2019] [Indexed: 02/08/2023] Open
Abstract
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising cancer therapeutic agent due to its selective killing on cancer cells while sparing the normal cells. Nevertheless, breast adenocarcinoma cells can develop TRAIL resistance. Therefore, this project investigated the anti-cancer effects of the combination of epigenetic drugs zebularine and trichostatin A (ZT) with TRAIL (TZT) on the human breast adenocarcinoma cells. This treatment regimen was compared with the natural anti-cancer compound curcumin (Cur) and standard chemotherapeutic drug doxorubicin (Dox). As compared to TRAIL treatment, TZT treatment hampered the cell viability of human breast adenocarcinoma cells MDA-MB-231 significantly but not MCF-7 and immortalized non-cancerous human breast epithelial cells MCF10A. Unlike TZT, Cur and Dox treatments reduced cell viability in both human breast adenocarcinoma and epithelial cells significantly. Nevertheless, there were no changes in cell cycle in both TRAIL and TZT treatments in breast adenocarcinoma and normal epithelial cells. Intriguingly, Cur and Dox treatment generally induced G2/M arrest in MDA-MB-231, MCF-7 and MCF10A but Cur induced S phase arrest in MCF10A. The features of apoptosis such as morphological changes, apoptotic activity and the expression of cleaved poly (ADP) ribose polymerase (PARP) protein were more prominent in TRAIL and TZT-treated MDA-MB-231 as compared to MCF10A at 24 h post-treatment. Compared to TZT treatment, Cur and Dox treatments exhibited lesser apoptotic features in MDA-MB-231. Collectively, the sensitization using Zeb and TSA to augment TRAIL-induced apoptosis might be an alternative therapy towards human breast adenocarcinoma cells, without harming the normal human breast epithelial cells.
Collapse
Affiliation(s)
- Wei Yang Kong
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, Semenyih, 43500, Malaysia
| | - Zong Yang Yee
- School of Post-Graduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur, 57000, Malaysia
| | - Chun Wai Mai
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, 57000, Malaysia
- Centre for Cancer and Stem Cell Research, Institute for Research, Development and Innovation, International Medical University, Bukit Jalil, Kuala Lumpur, 57000, Malaysia
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, Semenyih, 43500, Malaysia
| | - Syahril Abdullah
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, 43400, Malaysia
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor, 43400, Malaysia
| | - Siew Ching Ngai
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, Semenyih, 43500, Malaysia
| |
Collapse
|
38
|
Fei HR, Yuan C, Wang GL, Zhao Y, Li ZJ, Du X, Wang FZ. Caudatin potentiates the anti-tumor effects of TRAIL against human breast cancer by upregulating DR5. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 62:152950. [PMID: 31102888 DOI: 10.1016/j.phymed.2019.152950] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 05/04/2019] [Accepted: 05/05/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND The ability of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to preferentially induce apoptosis in transformed cells while sparing most normal cells is well established. However, the intrinsic and acquired resistance of tumors to TRAIL-induced apoptosis limits its therapeutic applicability. PURPOSE We investigated the effect of caudatin, a species of C-21 steroidal glycosides isolated from the roots of Cynanchum auriculatum, on TRAIL-induced apoptosis in human breast cancer cells. METHODS Cell growth inhibition was evaluated by the CCK-8 assay. The cell cycle distribution was assessed by propidium iodide flow cytometry. Apoptosis was determined by TUNEL staining. Protein expression was detected by western blotting analysis. RESULTS Caudatin enhanced TRAIL-induced apoptosis in human breast cancer cells. This sensitization was achieved by upregulating death receptor 5 (DR5). Knockdown of DR5 abolished the enhancing effect of caudatin on TRAIL responses. The caudatin-induced upregulation of DR5 was accompanied by increased expression of CHOP and phosphorylation of p38 MAPK and JNK. CHOP knockdown blocked caudatin-upregulated DR5 expression. Moreover, cotreatment of breast cancer cells with p38 MAPK and JNK inhibitors significantly counteracted the caudatin-induced expression of DR5. CONCLUSION Our results showed that caudatin sensitized breast cancer cells to TRAIL-induced apoptosis through activation of CHOP, p38 MAPK and JNK-mediated upregulation of DR5 expression. The combination of TRAIL and caudatin may be a promising therapeutic approach for the treatment of breast cancer.
Collapse
Affiliation(s)
- Hong-Rong Fei
- School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China
| | - Chuang Yuan
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China
| | - Gui-Ling Wang
- School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China
| | - Ying Zhao
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China
| | - Zhao-Jun Li
- School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China; School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China
| | - Xin Du
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China
| | - Feng-Ze Wang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China.
| |
Collapse
|
39
|
Lim B, Greer Y, Lipkowitz S, Takebe N. Novel Apoptosis-Inducing Agents for the Treatment of Cancer, a New Arsenal in the Toolbox. Cancers (Basel) 2019; 11:cancers11081087. [PMID: 31370269 PMCID: PMC6721450 DOI: 10.3390/cancers11081087] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/11/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023] Open
Abstract
Evasion from apoptosis is an important hallmark of cancer cells. Alterations of apoptosis pathways are especially critical as they confer resistance to conventional anti-cancer therapeutics, e.g., chemotherapy, radiotherapy, and targeted therapeutics. Thus, successful induction of apoptosis using novel therapeutics may be a key strategy for preventing recurrence and metastasis. Inhibitors of anti-apoptotic molecules and enhancers of pro-apoptotic molecules are being actively developed for hematologic malignancies and solid tumors in particular over the last decade. However, due to the complicated apoptosis process caused by a multifaceted connection with cross-talk pathways, protein–protein interaction, and diverse resistance mechanisms, drug development within the category has been extremely challenging. Careful design and development of clinical trials incorporating predictive biomarkers along with novel apoptosis-inducing agents based on rational combination strategies are needed to ensure the successful development of these molecules. Here, we review the landscape of currently available direct apoptosis-targeting agents in clinical development for cancer treatment and update the related biomarker advancement to detect and validate the efficacy of apoptosis-targeted therapies, along with strategies to combine them with other agents.
Collapse
Affiliation(s)
- Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Yoshimi Greer
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Stanley Lipkowitz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Naoko Takebe
- Early Clinical Trials Development, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
40
|
Wu N, Huang J, Zhang XF, Ou-Yang L, He S, Zhu Z, Xie W. Weighted Fused Pathway Graphical Lasso for Joint Estimation of Multiple Gene Networks. Front Genet 2019; 10:623. [PMID: 31396259 PMCID: PMC6662592 DOI: 10.3389/fgene.2019.00623] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/13/2019] [Indexed: 01/17/2023] Open
Abstract
Gene regulatory networks (GRNs) are often inferred based on Gaussian graphical models that could identify the conditional dependence among genes by estimating the corresponding precision matrix. Classical Gaussian graphical models are usually designed for single network estimation and ignore existing knowledge such as pathway information. Therefore, they can neither make use of the common information shared by multiple networks, nor can they utilize useful prior information to guide the estimation. In this paper, we propose a new weighted fused pathway graphical lasso (WFPGL) to jointly estimate multiple networks by incorporating prior knowledge derived from known pathways and gene interactions. Based on the assumption that two genes are less likely to be connected if they do not participate together in any pathways, a pathway-based constraint is considered in our model. Moreover, we introduce a weighted fused lasso penalty in our model to take into account prior gene interaction data and common information shared by multiple networks. Our model is optimized based on the alternating direction method of multipliers (ADMM). Experiments on synthetic data demonstrate that our method outperforms other five state-of-the-art graphical models. We then apply our model to two real datasets. Hub genes in our identified state-specific networks show some shared and specific patterns, which indicates the efficiency of our model in revealing the underlying mechanisms of complex diseases.
Collapse
Affiliation(s)
- Nuosi Wu
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, China
| | - Jiang Huang
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, China
| | - Xiao-Fei Zhang
- School of Mathematics and Statistics, Central China Normal University, Wuhan, China
| | - Le Ou-Yang
- Guangdong Key Laboratory of Intelligent Information Processing and Shenzhen Key Laboratory of Media Security, College of Electronics and Information Engineering, Shenzhen University, Shenzhen, China
- Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen, China
| | - Shan He
- School of Computer Science, University of Birmingham, Birmingham, United Kingdom
| | - Zexuan Zhu
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, China
| | - Weixin Xie
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
41
|
Jeon MY, Seo SU, Woo SM, Min KJ, Byun HS, Hur GM, Kang SC, Kwon TK. Oridonin enhances TRAIL-induced apoptosis through GALNT14-mediated DR5 glycosylation. Biochimie 2019; 165:108-114. [PMID: 31336136 DOI: 10.1016/j.biochi.2019.07.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 07/17/2019] [Indexed: 01/09/2023]
Abstract
Oridonin is a diterpenoid isolated from the Rabdosia rubescens and has multiple biological effects, such as anti-inflammation and anti-tumor activities. In present study, we revealed that the sensitizing effect of oridonin on tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in several cancer cells, but not in normal cells. Oridonin enhanced death-signaling inducing complexes (DISC) formation and DR5 glycosylation without affecting expression of downstream intracellular apoptosis-related proteins. Oridonin upregulated peptidyl O-glycosyltransferase GALNT14 in a dose- and time-dependent manner. Knockdown of GALNT14 by siRNA and Endo H treatment reduced oridonin-induced DR5 glycosylation. Furthermore, treatment with inhibitor of glycosylation (benzyl-α-GalNAc) blocked oridonin plus TRAIL-induced apoptosis. Collectively, our results suggest that oridonin-induced DR5 glycosylation contributes to TRAIL-induced apoptotic cell death in cancer cells.
Collapse
Affiliation(s)
- Mi-Yeon Jeon
- Department of Immunology, School of Medicine, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu, 42601, South Korea
| | - Seung Un Seo
- Department of Immunology, School of Medicine, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu, 42601, South Korea
| | - Seon Min Woo
- Department of Immunology, School of Medicine, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu, 42601, South Korea
| | - Kyoung-Jin Min
- Department of Immunology, School of Medicine, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu, 42601, South Korea
| | - Hee Sun Byun
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon, 35015, South Korea
| | - Gang Min Hur
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon, 35015, South Korea
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, South Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu, 42601, South Korea.
| |
Collapse
|
42
|
Pandey P, Singh D, Hasanain M, Ashraf R, Maheshwari M, Choyal K, Singh A, Datta D, Kumar B, Sarkar J. 7-hydroxyfrullanolide, isolated from Sphaeranthus indicus, inhibits colorectal cancer cell growth by p53-dependent and -independent mechanism. Carcinogenesis 2019; 40:791-804. [PMID: 30535334 DOI: 10.1093/carcin/bgy176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 11/06/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022] Open
Abstract
Sphaeranthus indicus Linn. is commonly used in Indian traditional medicine for management of multiple pathological conditions. However, there are limited studies on anticancer activity of this plant and its underlying molecular mechanisms. Here, we isolated an active constituent, 7-hydroxyfrullanolide (7-HF), from the flowers of this plant, which showed promising chemotherapeutic potential. The compound was more effective in inhibiting in vitro proliferation of colon cancers cells through G2/M phase arrest than other cancer cell lines that were used in this study. Consistent with in vitro data, 7-HF caused substantial regression of tumour volume in a syngeneic mouse model of colon cancer. The molecule triggered extrinsic apoptotic pathway, which was evident as upregulation of DR4 and DR5 expression as well as induction of their downstream effector molecules (FADD, Caspase-8). Concurrent activation of intrinsic pathway was demonstrated with loss of ΔΨm to release pro-apoptotic cytochrome c from mitochondria and activation of downstream caspase cascades (Caspase -9, -3). Loss of p53 resulted in decreased sensitivity of cells towards pro-apoptotic effect of 7-HF with increased number of viable cells indicating p53-dependent arrest of cancer cell growth. This notion was further supported with 7-HF-mediated elevation of endogenous p53 level, decreased expression of MDM2 and transcriptional upregulation of p53 target genes in apoptotic pathway. However, 7-HF was equally effective in preventing progression of HCT116 p53+/+ and p53-/- cell derived xenografts in nude mice, which suggests that differences in p53 status may not influence its in vivo efficacy. Taken together, our results support 7-HF as a potential chemotherapeutic agent and provided a new mechanistic insight into its anticancer activity.
Collapse
Affiliation(s)
- Praveen Pandey
- Biochemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, Uttar Pradesh, India
| | - Deepika Singh
- Sophisticated Analytical Instrument Facility, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, Uttar Pradesh, India
| | - Mohammad Hasanain
- Biochemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India
| | - Raghib Ashraf
- Biochemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, Uttar Pradesh, India
| | - Mayank Maheshwari
- Biochemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, Uttar Pradesh, India
| | - Kuldeep Choyal
- Biochemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, Uttar Pradesh, India
| | - Akhilesh Singh
- Biochemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, Uttar Pradesh, India
| | - Dipak Datta
- Biochemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India
| | - Brijesh Kumar
- Sophisticated Analytical Instrument Facility, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India
| | - Jayanta Sarkar
- Biochemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India.,Laboratory Animal Facility, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, Uttar Pradesh, India
| |
Collapse
|
43
|
Twomey JD, Zhao L, Luo S, Xu Q, Zhang B. Tubulin couples death receptor 5 to regulate apoptosis. Oncotarget 2018; 9:36804-36815. [PMID: 30613368 PMCID: PMC6298406 DOI: 10.18632/oncotarget.26407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 11/16/2018] [Indexed: 12/30/2022] Open
Abstract
Activation of death receptor 5 (DR5) to induce apoptosis in cancer cells is an attractive strategy for cancer therapy. However, many tumor cell lines and primary tumors are resistant to DR5 targeted agents including recombinant tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and anti-DR5 agonistic antibodies. Here we identify tubulin proteins - primarily consisting of α and β subunits folded into microtubule polymers - as a crucial modulator of DR5 mediated apoptosis. Using affinity purification coupled with mass spectrometry, we found that DR5 interacts with both α- and β-tubulin proteins in cancer cells. Pharmacological disruption of microtubules increased DR5 protein expression and subsequently sensitized the cells to TRAIL-induced apoptosis. Similar results were observed by selectively silencing tubulin transcript using small RNA interference. We also demonstrate that tubulin/microtubule blockade augments TRAIL induced apoptosis by stabilizing DR5 protein. Together, our results link the tubulin/microtubule network to the stringent regulation of DR5 mediated apoptosis, which could lead to potential therapeutic strategies to enhance cancer therapy efficacy.
Collapse
Affiliation(s)
- Julianne D Twomey
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Liqun Zhao
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Shen Luo
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Qing Xu
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Baolin Zhang
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
44
|
Turner KA, Manouchehri JM, Kalafatis M. Sensitization of recombinant human tumor necrosis factor-related apoptosis-inducing ligand-resistant malignant melanomas by quercetin. Melanoma Res 2018; 28:277-285. [PMID: 29596115 PMCID: PMC6039425 DOI: 10.1097/cmr.0000000000000447] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 02/20/2018] [Indexed: 12/30/2022]
Abstract
Malignant melanoma is the most commonly diagnosed skin cancer associated with a high rate of metastasis. Low-stage melanoma is easily treated, but metastatic malignant melanoma is an extremely treatment-resistant malignancy with low survival rates. The application of recombinant human tumor necrosis factor-related apoptosis-inducing ligand (rhTRAIL) for the treatment of metastatic malignant melanoma holds considerable promise because of its selective proapoptotic activity towards cancer cells and not nontransformed cells. Unfortunately, the clinical utilization of rhTRAIL has been terminated due to the resistance of many cancer cells to undergo apoptosis in response to rhTRAIL. However, rhTRAIL-resistance can be abrogated through the cotreatment with compounds derived from 'Mother Nature' such as quercetin that can modulate cellular components responsible for rhTRAIL-resistance. Here, we show that rhTRAIL-resistant malignant melanomas are sensitized by quercetin. Quercetin action is manifested by the upregulation of rhTRAIL-binding receptors DR4 and DR5 on the surface of cancer cells and by increased rate of the proteasome-mediated degradation of the antiapoptotic protein FLIP. Our data provide for a new efficient and nontoxic treatment of malignant melanoma.
Collapse
Affiliation(s)
- Katherine A. Turner
- Department of Chemistry, Cleveland State University
- Center for Gene Regulation in Health and Disease (GRHD)
| | - Jasmine M. Manouchehri
- Department of Chemistry, Cleveland State University
- Center for Gene Regulation in Health and Disease (GRHD)
| | - Michael Kalafatis
- Department of Chemistry, Cleveland State University
- Center for Gene Regulation in Health and Disease (GRHD)
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
45
|
Alimbetov D, Askarova S, Umbayev B, Davis T, Kipling D. Pharmacological Targeting of Cell Cycle, Apoptotic and Cell Adhesion Signaling Pathways Implicated in Chemoresistance of Cancer Cells. Int J Mol Sci 2018; 19:ijms19061690. [PMID: 29882812 PMCID: PMC6032165 DOI: 10.3390/ijms19061690] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/11/2022] Open
Abstract
Chemotherapeutic drugs target a physiological differentiating feature of cancer cells as they tend to actively proliferate more than normal cells. They have well-known side-effects resulting from the death of highly proliferative normal cells in the gut and immune system. Cancer treatment has changed dramatically over the years owing to rapid advances in oncology research. Developments in cancer therapies, namely surgery, radiotherapy, cytotoxic chemotherapy and selective treatment methods due to better understanding of tumor characteristics, have significantly increased cancer survival. However, many chemotherapeutic regimes still fail, with 90% of the drug failures in metastatic cancer treatment due to chemoresistance, as cancer cells eventually develop resistance to chemotherapeutic drugs. Chemoresistance is caused through genetic mutations in various proteins involved in cellular mechanisms such as cell cycle, apoptosis and cell adhesion, and targeting those mechanisms could improve outcomes of cancer therapy. Recent developments in cancer treatment are focused on combination therapy, whereby cells are sensitized to chemotherapeutic agents using inhibitors of target pathways inducing chemoresistance thus, hopefully, overcoming the problems of drug resistance. In this review, we discuss the role of cell cycle, apoptosis and cell adhesion in cancer chemoresistance mechanisms, possible drugs to target these pathways and, thus, novel therapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Dauren Alimbetov
- Laboratory of bioengineering and regenerative medicine, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave, Z05H0P9 Astana, Kazakhstan.
| | - Sholpan Askarova
- Laboratory of bioengineering and regenerative medicine, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave, Z05H0P9 Astana, Kazakhstan.
| | - Bauyrzhan Umbayev
- Laboratory of bioengineering and regenerative medicine, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave, Z05H0P9 Astana, Kazakhstan.
| | - Terence Davis
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK.
| | - David Kipling
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
46
|
Yano S, Wu S, Sakao K, Hou DX. Wasabi 6-(methylsulfinyl)hexyl isothiocyanate induces apoptosis in human colorectal cancer cells through p53-independent mitochondrial dysfunction pathway. Biofactors 2018; 44:361-368. [PMID: 29756671 DOI: 10.1002/biof.1431] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/30/2018] [Accepted: 04/13/2018] [Indexed: 12/18/2022]
Abstract
6-(Methylsulfinyl)hexyl isothiocyanate (6-MSITC), a major bioactive compound in Wasabi [Wasabia japonica (Miq.) Matsum.], has revealed the inhibitory effect on colon carcinogenesis in rat cancer model although the underlying mechanism is unclear. In this study, we used two types of human colorectal cancer cells (HCT116 p53+/+ and HCT116 p53-/- ) to investigate the anticancer activity and molecular mechanisms of 6-MSITC. Interestingly, 6-MSITC inhibited the cell proliferation in both types of cells with similar IC50 value although a light increase in the phosphorylation and accumulation of P53 protein was observed in HCT116 p53+/+ cells at 24 h after treatment. In addition, 6-MSITC increased the ratio of proapoptotic cells in both types of cells with the same fashion in a p53-independent manner. The data from mitochondrial analysis revealed that 6-MSITC enhanced the ratio of proapoptotic B-cell lymphoma-2-associated X protein/antiapoptotic myeloid cell leukemia 1, and sequentially caused mitochondrial membrane potential (ΔΨm ) loss, cytochrome c release, and caspase-3 activation in both types of cells. Taken together, Wasabi 6-MSITC induced apoptosis of human colorectal cancer cells in p53-independent mitochondrial dysfunction pathway. These findings suggest that 6-MSITC might be a potential agent for colon cancer chemoprevention although with p53 mutation. © 2018 BioFactors, 2018.
Collapse
Affiliation(s)
- Satoshi Yano
- Course of Biological Science and Technology, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Shusong Wu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Kozue Sakao
- Course of Biological Science and Technology, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
- Department of Food Science and Biotechnology, Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
| | - De-Xing Hou
- Course of Biological Science and Technology, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
- Department of Food Science and Biotechnology, Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
47
|
Wang X, Xue Q, Wu L, Wang B, Liang H. Dasatinib promotes TRAIL-mediated apoptosis by upregulating CHOP-dependent death receptor 5 in gastric cancer. FEBS Open Bio 2018; 8:732-742. [PMID: 29744288 PMCID: PMC5929929 DOI: 10.1002/2211-5463.12404] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/21/2018] [Accepted: 02/02/2018] [Indexed: 12/12/2022] Open
Abstract
Dasatinib, a tyrosine kinase inhibitor, has been approved for first‐line treatment of leukemia and has also been evaluated for use in numerous other cancers. However, its role in gastric cancer (GC) remains unclear. Therefore, the aim of this study was to investigate how dasatinib suppresses the growth of GC cells and interacts with chemotherapeutic drugs. The results showed that, in the presence of dasatinib, proliferation of GC cells decreased and apoptosis increased, and that Fas‐associated death domain protein and caspase‐8 are essential to dasatinib‐induced cell apoptosis in GC. In addition, we found that dasatinib increased the expression of death receptor 5 (DR5) in GC cells. Dasatinib enhanced apoptosis induced by tumor necrosis factor‐related apoptosis‐inducing ligand (TRAIL) in GC cells. Moreover, increased DR5 expression facilitated dasatinib‐induced apoptosis; the dasatinib‐induced increase in DR5 expression was mediated by CCAAT/enhancer‐binding protein homologous protein (CHOP). Furthermore, dasatinib also synergized with TRAIL to induce apoptosis via DR5 in GC cells. Our results show that dasatinib promoted TRAIL‐mediated apoptosis via upregulation of CHOP‐dependent DR5 expression in GC, suggesting that DR5 induction can be used as an indicator of dasatinib sensitivity.
Collapse
Affiliation(s)
- Xiaona Wang
- Department of Gastric Cancer Tianjin Medical University Cancer Institute and Hospital National Clinical Research Center for Cancer Key Laboratory of Cancer Prevention and Therapy Tianjin's Clinical Research Center for Cancer China
| | - Qiang Xue
- Department of Gastric Cancer Tianjin Medical University Cancer Institute and Hospital National Clinical Research Center for Cancer Key Laboratory of Cancer Prevention and Therapy Tianjin's Clinical Research Center for Cancer China
| | - Liangliang Wu
- Department of Gastric Cancer Tianjin Medical University Cancer Institute and Hospital National Clinical Research Center for Cancer Key Laboratory of Cancer Prevention and Therapy Tianjin's Clinical Research Center for Cancer China
| | - Baogui Wang
- Department of Gastric Cancer Tianjin Medical University Cancer Institute and Hospital National Clinical Research Center for Cancer Key Laboratory of Cancer Prevention and Therapy Tianjin's Clinical Research Center for Cancer China
| | - Han Liang
- Department of Gastric Cancer Tianjin Medical University Cancer Institute and Hospital National Clinical Research Center for Cancer Key Laboratory of Cancer Prevention and Therapy Tianjin's Clinical Research Center for Cancer China
| |
Collapse
|
48
|
Oh YT, Qian G, Deng J, Sun SY. Monocyte chemotactic protein-induced protein-1 enhances DR5 degradation and negatively regulates DR5 activation-induced apoptosis through its deubiquitinase function. Oncogene 2018; 37:3415-3425. [PMID: 29551769 DOI: 10.1038/s41388-018-0200-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 12/20/2017] [Accepted: 02/09/2018] [Indexed: 11/10/2022]
Abstract
Monocyte chemotactic protein-induced protein-1 (MCPIP1; also called Regnase-1) encoded by the ZC3H12A gene critically regulates inflammatory responses and immune homeostasis primarily by RNase-dependent and -independent mechanisms. However, the relationship of MCPIP1 with apoptosis and cancer and the underlying mechanisms are largely unclear. The current study has demonstrated a previously uncovered connection between MCPIP1 and the negative regulation of death receptor 5 (DR5; also known as TRAIL-R2 or killer/DR5), a cell surface receptor for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), which is produced endogenously by various immune cells such as T cells. Our findings have revealed that MCPIP1 decreases both total cellular and cell surface DR5, primarily through modulating DUB-mediated protein autophagic/lysosomal degradation. Suppression of MCPIP1 by gene knockdown induces the formation of death-induced signaling complex (DISC) and enhances TRAIL or DR5 activation-induced apoptosis in cancer cells. Moreover, we demonstrated an inverse correlation between MCPIP1 expression and DR5 expression/cell sensitivity to DR5 activation-induced apoptosis in cancer cells. Our findings warrant future investigation of the roles of negative regulation of DR5 by MCPIP1 in cancer and in T-cell immunity.
Collapse
Affiliation(s)
- You-Take Oh
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Guoqing Qian
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Jiusheng Deng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
49
|
Das S, Nayak A, Siddharth S, Nayak D, Narayan S, Kundu CN. TRAIL enhances quinacrine-mediated apoptosis in breast cancer cells through induction of autophagy via modulation of p21 and DR5 interactions. Cell Oncol (Dordr) 2017; 40:593-607. [DOI: 10.1007/s13402-017-0347-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2017] [Indexed: 01/20/2023] Open
|
50
|
Pan ST, Li ZL, He ZX, Qiu JX, Zhou SF. Molecular mechanisms for tumour resistance to chemotherapy. Clin Exp Pharmacol Physiol 2017; 43:723-37. [PMID: 27097837 DOI: 10.1111/1440-1681.12581] [Citation(s) in RCA: 259] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 04/13/2016] [Accepted: 04/18/2016] [Indexed: 12/25/2022]
Abstract
Chemotherapy is one of the prevailing methods used to treat malignant tumours, but the outcome and prognosis of tumour patients are not optimistic. Cancer cells gradually generate resistance to almost all chemotherapeutic drugs via a variety of distinct mechanisms and pathways. Chemotherapeutic resistance, either intrinsic or acquired, is caused and sustained by reduced drug accumulation and increased drug export, alterations in drug targets and signalling transduction molecules, increased repair of drug-induced DNA damage, and evasion of apoptosis. In order to better understand the mechanisms of chemoresistance, this review highlights our current knowledge of the role of altered drug metabolism and transport and deregulation of apoptosis and autophagy in the development of tumour chemoresistance. Reduced intracellular activation of prodrugs (e.g. thiotepa and tegafur) or enhanced drug inactivation by Phase I and II enzymes contributes to the development of chemoresistance. Both primary and acquired resistance can be caused by alterations in the transport of anticancer drugs which is mediated by a variety of drug transporters such as P-glycoprotein (P-gp), multidrug resistance associated proteins, and breast cancer resistance protein. Presently there is a line of evidence indicating that deregulation of programmed cell death including apoptosis and autophagy is also an important mechanism for tumour resistance to anticancer drugs. Reversal of chemoresistance is likely via pharmacological and biological approaches. Further studies are warranted to grasp the full picture of how each type of cancer cells develop resistance to anticancer drugs and to identify novel strategies to overcome it.
Collapse
Affiliation(s)
- Shu-Ting Pan
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Zhi-Ling Li
- Department of Pharmacy, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Xu He
- Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Centre & Sino-US Joint Laboratory for Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jia-Xuan Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| |
Collapse
|