1
|
Jung HJ, Kim JK, Suh SI, Baek WK. Chaetocin enhances tumor necrosis factor‑related apoptosis‑inducing ligand‑mediated apoptosis by enhancing DR5 stabilization and reactive oxygen species generation in human glioblastoma cells. Int J Oncol 2025; 66:47. [PMID: 40377008 PMCID: PMC12118949 DOI: 10.3892/ijo.2025.5753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 03/05/2025] [Indexed: 05/18/2025] Open
Abstract
Chaetocin, a fungal metabolite, exerts notable antiproliferative effects against solid tumors by triggering apoptosis; however, the mechanisms underlying its effects remain unclear. As tumor necrosis factor (TNF)‑related apoptosis‑inducing ligand (TRAIL) promotes apoptosis in certain types of tumor, the present study aimed to explore the sensitizing effects of chaetocin in TRAIL‑induced apoptosis in human glioblastoma cells and the underlying mechanism. Human glioblastoma cells (U343MG, U87MG, U251MG, and T98G) and embryonic kidney cells (HEK293) were co‑treated with chaetocin and TRAIL, followed by assessment of cell viability. The results from viability and apoptosis assays demonstrated a significant increase in caspase-dependent apoptosis in glioblastoma cells, but not in HEK293 cells, upon co-treatment with chaetocin and TRAIL. Additionally, death receptor 5 (DR5) expression analysis demonstrated that co‑treatment with chaetocin and TRAIL upregulated DR5 expression in a dose‑ and time‑dependent manner by increasing the stability of DR5 on the cell surface. In glioblastoma cells, small interfering RNA‑mediated DR5 knockdown markedly suppressed chaetocin/TRAIL‑induced apoptosis. Moreover, chaetocin enhanced reactive oxygen species (ROS) production, which facilitated TRAIL‑mediated apoptosis by enhancing DR5 upregulation. Thus, chaetocin sensitized the human glioblastoma cell lines U87MG and T98G to TRAIL‑mediated apoptosis by upregulating DR5 expression through ROS-mediated mechanisms. The present findings underscore chaetocin as a potential novel therapeutic agent for glioblastoma.
Collapse
Affiliation(s)
- Hui-Jung Jung
- Department of Microbiology, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Seong-Il Suh
- Department of Microbiology, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Won-Ki Baek
- Department of Microbiology, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
- Institute for Cancer Research, Keimyung University Dongsan Medical Center, Dalseogu, Daegu 42601, Republic of Korea
| |
Collapse
|
2
|
Gao H, Ma B, Zhao Y, Pan Y, Zhang A. Implications and mechanisms of O-GlcNAcylation in cancer therapy resistance. TUMORI JOURNAL 2025; 111:41-54. [PMID: 39718082 DOI: 10.1177/03008916241299244] [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] [Indexed: 12/25/2024]
Abstract
O-linked-N-acetylglucosaminylation (O-GlcNAcylation), one of the protein post-translational modifications, is the process of adding O-linked-β-D-N-acetylglucosaminylation (O-GlcNAc) to serine and threonine residues of proteins. O-GlcNAcylation regulates various fundamental cell biological processes, including gene transcription, signal transduction, and cellular metabolism. The role of dysregulated O-GlcNAcylation in tumorigenesis has been recognized, but its role in cancer therapy tolerance has not been elucidated. Therefore, this paper provides the latest evidence on the role of O-GlcNAcylation in cancer therapy responsiveness to understand the impact of O-GlcNAcylation on cancer therapy outcomes, as well as analyzing several possible mechanisms by which O-GlcNAcylation dysregulation affects cancer therapy efficacy, and discusses the possibility of O-GlcNAcylation as a cancer therapy sensitizer.
Collapse
Affiliation(s)
- Hongwei Gao
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Binyuan Ma
- Healthy Examination & Management Center, First Hospital of Lanzhou University, Lanzhou, China
| | - Youli Zhao
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yunyan Pan
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Anan Zhang
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| |
Collapse
|
3
|
Huang D, Yang X, Peng Z, Yin H, Liu Y, Zhang Y, Li C, Chen G, Wang Q. Multichannel-optical imaging for in vivo evaluating the safety and therapeutic efficacy of stem cells in tumor model in terms of cell tropism, proliferation and NF-κB activity. Biomaterials 2024; 307:122510. [PMID: 38422837 DOI: 10.1016/j.biomaterials.2024.122510] [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: 09/22/2023] [Revised: 01/20/2024] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
Stem cell-based cancer treatment has garnered significant attention, yet its safety and efficacy remain incompletely understood. The nuclear factor-kappa B (NF-κB) pathway, a critical signaling mechanism involved in tumor growth, angiogenesis, and invasion, serves as an essential metric for evaluating the behavior of stem cells in tumor models. Herein, we report the development of a triple-channel imaging system capable of simultaneously monitoring the tropism of stem cells towards tumors, assessing tumor proliferation, and quantifying tumor NF-κB activity. In this system, we generated a CRISPR-Cas9 gene-edited human glioblastoma cell line, GE-U87-MG, which provided a reliable readout of the proliferation and NF-κB activity of tumors by EF1α-RFLuc- and NF-κB-GLuc-based bioluminescent imaging, respectively. Additionally, near infrared-II emitting Tat-PEG-AgAuSe quantum dots were developed for tracking of stem cell tropism towards tumor. In a representative case involving human mesenchymal stem cells (hMSCs), multichannel imaging revealed no discernible effect of hMSCs on the proliferation and NF-κB activity of GE-U87-MG tumors. Moreover, hMSCs engineered to overexpress the necrosis factor-related apoptosis-inducing ligand were able to inhibit NF-κB activity and growth of GE-U87-MG in vivo. Taken together, our imaging system represents a powerful and feasible approach to evaluating the safety and therapeutic efficacy of stem cells in tumor models.
Collapse
Affiliation(s)
- Dehua Huang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Xue Yang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zhao Peng
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Hongqiang Yin
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Yongyang Liu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Yejun Zhang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Chunyan Li
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Guangcun Chen
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China.
| | - Qiangbin Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
4
|
Kahramanoğullari M, Erişir M, Yaman M, Parlak Ak T. Effects of naringenin on oxidative damage and apoptosis in liver and kidney in rats subjected to chronic mercury chloride. ENVIRONMENTAL TOXICOLOGY 2024; 39:2937-2947. [PMID: 38308452 DOI: 10.1002/tox.24164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/03/2024] [Accepted: 01/18/2024] [Indexed: 02/04/2024]
Abstract
Mercury chloride is a type of heavy metal that causes the formation of free radicals, causing hepatotoxicity, nephrotoxicity and apoptosis. In this study, the effects of naringenin on oxidative stress and apoptosis in the liver and kidney of rats exposed to mercury chloride were investigated. In the study, 41 2-month-old male Wistar-Albino rats were divided into five groups. Accordingly, group 1 was set as control group, group 2 as naringenin-100, group 3 as mercury chloride, group 4 as mercury chloride + naringenin-50, and group 5 as mercury chloride + naringenin-100. For the interventions, 1 mL/kg saline was administered to the control, 0.4 mg/kg/day mercury (II) chloride to the mercury chloride groups by i.p., and 50 and 100 mg/kg/day naringenin prepared in corn oil to the naringenin groups by gavage. All the interventions lasted for 20 days. Mercury chloride administration was initiated 1 h following the administration of naringenin. When mercury chloride and the control group were compared, a significant increase in plasma urea, liver and kidney malondialdehyde (MDA) levels, in kidney superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), glutathione-S-transferase (GST) activities (p < .001), and a significant decrease in liver and kidney glutathione (GSH) levels (p < .001), in liver catalase (CAT) activity (p < .01) were observed. In addition, histopathological changes and a significant increase in caspase-3 levels were detected (p < .05). When mercury chloride and treatment groups were compared, the administration of naringenin caused a decrease aspartate transaminase (AST), alanine transaminase (ALT), lactate dehydrogenase (LDH) (p < .01), urea, creatinine levels (p < .001) in plasma, MDA levels in liver and kidney, SOD, GSH-Px, GST activities in kidney (p < .001), and increased GSH levels in liver and kidney. The addition of naringenin-100 increased GSH levels above the control (p < .001). The administration of naringenin was also decreased histopathological changes and caspase-3 levels (p < .05). Accordingly, it was determined that naringenin is protective and therapeutic against mercury chloride-induced oxidative damage and apoptosis in the liver and kidney, and 100 mg/kg naringenin is more effective in preventing histopathological changes and apoptosis.
Collapse
Affiliation(s)
- Merve Kahramanoğullari
- Department of Biochemistry, Faculty of Veterinary Medicine, Fırat University, Elazığ, Turkey
| | - Mine Erişir
- Department of Biochemistry, Faculty of Veterinary Medicine, Fırat University, Elazığ, Turkey
| | - Mine Yaman
- Department of Histology-Embryology, Faculty of Veterinary Medicine, Fırat University, Elazığ, Turkey
| | - Tuba Parlak Ak
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Munzur University, Tunceli, Turkey
| |
Collapse
|
5
|
Davidson BA, Miranda AX, Reed SC, Bergman RE, Kemp JDJ, Reddy AP, Pantone MV, Fox EK, Dorand RD, Hurley PJ, Croessmann S, Park BH. An in vitro CRISPR screen of cell-free DNA identifies apoptosis as the primary mediator of cell-free DNA release. Commun Biol 2024; 7:441. [PMID: 38600351 PMCID: PMC11006667 DOI: 10.1038/s42003-024-06129-1] [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: 08/22/2023] [Accepted: 03/29/2024] [Indexed: 04/12/2024] Open
Abstract
ABTRACT Clinical circulating cell-free DNA (cfDNA) testing is now routine, however test accuracy remains limited. By understanding the life-cycle of cfDNA, we might identify opportunities to increase test performance. Here, we profile cfDNA release across a 24-cell line panel and utilize a cell-free CRISPR screen (cfCRISPR) to identify mediators of cfDNA release. Our panel outlines two distinct groups of cell lines: one which releases cfDNA fragmented similarly to clinical samples and purported as characteristic of apoptosis, and another which releases larger fragments associated with vesicular or necrotic DNA. Our cfCRISPR screens reveal that genes mediating cfDNA release are primarily involved with apoptosis, but also identify other subsets of genes such as RNA binding proteins as potential regulators of cfDNA release. We observe that both groups of cells lines identified primarily produce cfDNA through apoptosis. These results establish the utility of cfCRISPR, genetically validate apoptosis as a major mediator of DNA release in vitro, and implicate ways to improve cfDNA assays.
Collapse
Affiliation(s)
- Brad A Davidson
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Adam X Miranda
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Sarah C Reed
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
| | - Riley E Bergman
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
| | - Justin D J Kemp
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Anvith P Reddy
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
| | - Morgan V Pantone
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Ethan K Fox
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - R Dixon Dorand
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Paula J Hurley
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Sarah Croessmann
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Ben Ho Park
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
| |
Collapse
|
6
|
Yu J, Li X, Cao J, Zhu T, Liang S, Du L, Cao M, Wang H, Zhang Y, Zhou Y, Shen B, Feng J, Zhang J, Wang J, Jin J. Components of the JNK-MAPK pathway play distinct roles in hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149:17495-17509. [PMID: 37902853 DOI: 10.1007/s00432-023-05473-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/10/2023] [Indexed: 11/01/2023]
Abstract
PURPOSE Mitogen-activated protein kinases (MAPK), specifically the c-Jun N-terminal kinase (JNK)-MAPK subfamily, play a crucial role in the development of various cancers, including hepatocellular carcinoma (HCC). However, the specific roles of JNK1/2 and their upstream regulators, MKK4/7, in HCC carcinogenesis remain unclear. METHODS In this study, we performed differential expression analysis of JNK-MAPK components at both the transcriptome and protein levels using TCGA and HPA databases. We utilized Kaplan-Meier survival plots and receiver operating characteristic (ROC) curve analysis to evaluate the prognostic performance of a risk scoring model based on these components in the TCGA-HCC cohort. Additionally, we conducted immunoblotting, apoptosis analysis with FACS and soft agar assays to investigate the response of JNK-MAPK pathway components to various death stimuli (TRAIL, TNF-α, anisomycin, and etoposide) in HCC cell lines. RESULTS JNK1/2 and MKK7 levels were significantly upregulated in HCC samples compared to paracarcinoma tissues, whereas MKK4 was downregulated. ROC analyses suggested that JNK2 and MKK7 may serve as suitable diagnostic genes for HCC, and high JNK2 expression correlated with significantly poorer overall survival. Knockdown of JNK1 enhanced TRAIL-induced apoptosis in hepatoma cells, while JNK2 knockdown reduced TNF-α/cycloheximide (CHX)-and anisomycin-induced apoptosis. Neither JNK1 nor JNK2 knockdown affected etoposide-induced apoptosis. Furthermore, MKK7 knockdown augmented TNF-α/CHX- and TRAIL-induced apoptosis and inhibited colony formation in hepatoma cells. CONCLUSION Targeting MKK7, rather than JNK1/2 or MKK4, may be a promising therapeutic strategy to inhibit the JNK-MAPK pathway in HCC therapy.
Collapse
Affiliation(s)
- Jijun Yu
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Xinying Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Junxia Cao
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ting Zhu
- Beijing No. 80 High School, Beijing, 100102, China
| | - Shuifeng Liang
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
| | - Le Du
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
| | - Meng Cao
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
| | - Haitao Wang
- Department of Hematology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100071, China
| | - Yaolin Zhang
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yinxi Zhou
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
| | - Beifen Shen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Jiyan Zhang
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
| | - Jing Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Jianfeng Jin
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China.
| |
Collapse
|
7
|
Quiroz-Reyes AG, Delgado-González P, Islas JF, Soto-Domínguez A, González-Villarreal CA, Padilla-Rivas GR, Garza-Treviño EN. Oxaliplatin Enhances the Apoptotic Effect of Mesenchymal Stem Cells, Delivering Soluble TRAIL in Chemoresistant Colorectal Cancer. Pharmaceuticals (Basel) 2023; 16:1448. [PMID: 37895919 PMCID: PMC10610062 DOI: 10.3390/ph16101448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/03/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
A key problem in colorectal cancer (CRC) is the development of resistance to current therapies due to the presence of cancer stem cells (CSC), which leads to poor prognosis. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a protein that activates apoptosis in cancer cells through union with TRAIL death receptors. Cell therapies as delivery systems can produce soluble TRAIL (sTRAIL) and full-length TRAIL (flTRAIL), showing a high capacity to produce apoptosis in vitro and in vivo assays. However, the apoptotic activity of TRAIL as monotherapy had limitations, so it is important to explore other ways to enhance susceptibility to TRAIL. This study evaluated the cytotoxic and proapoptotic activity of soluble TRAIL overexpressed by mesenchymal stem cells (MSC) in an oxaliplatin-resistant CRC cell line. Bone marrow-MSC were lentiviral transduced for soluble TRAIL expression. DR5 death receptor expression was determined in Caco-2 and CMT-93 CRC cell lines. Sensitivity to first-line chemotherapies and recombinant TRAIL was evaluated by half-maximal inhibitory concentrations. Cytotoxic and proapoptotic activity of soluble TRAIL-MSC alone and combined with chemotherapy pre-treatment was evaluated using co-cultures. Caco-2 and CMT-93 cell lines expressed 59.08 ± 5.071 and 51.65 ± 11.99 of DR5 receptor and had IC50 of 534.15 ng/mL and 581.34 ng/mL for recombinant murine TRAIL (rmTRAIL), respectively. This finding was classified as moderate resistance to TRAIL. The Caco-2 cell line showed resistance to oxaliplatin and irinotecan. MSC successfully overexpressed soluble TRAIL and induced cancer cell death at a 1:6 ratio in co-culture. Oxaliplatin pre-treatment in the Caco-2 cell line increased the cell death percentage (50%) and apoptosis by sTRAIL. This finding was statistically different from the negative control (p < 0.05), and activity was even higher with the oxaliplatin-flTRAIL combination. Thus, oxaliplatin increases apoptotic activity induced by soluble TRAIL in a chemoresistant CRC cell line.
Collapse
Affiliation(s)
- Adriana G Quiroz-Reyes
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon, Monterrey 81, Mexico; (A.G.Q.-R.); (P.D.-G.); (J.F.I.); (G.R.P.-R.)
| | - Paulina Delgado-González
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon, Monterrey 81, Mexico; (A.G.Q.-R.); (P.D.-G.); (J.F.I.); (G.R.P.-R.)
| | - José F. Islas
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon, Monterrey 81, Mexico; (A.G.Q.-R.); (P.D.-G.); (J.F.I.); (G.R.P.-R.)
| | - Adolfo Soto-Domínguez
- Department of Histology, School of Medicine, Autonomous University of Nuevo Leon, Monterrey 81, Mexico;
| | | | - Gerardo R. Padilla-Rivas
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon, Monterrey 81, Mexico; (A.G.Q.-R.); (P.D.-G.); (J.F.I.); (G.R.P.-R.)
| | - Elsa N. Garza-Treviño
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon, Monterrey 81, Mexico; (A.G.Q.-R.); (P.D.-G.); (J.F.I.); (G.R.P.-R.)
| |
Collapse
|
8
|
Zhao D, Yang L, Han P, Zhang H, Wang F, Meng Z, Gan H, Wu Z, Sun W, Chen C, Dou G, Gu R. Blocking TRAIL-DR5 signaling pathway with soluble death receptor 5 fusion protein mitigates radiation-induced injury. Front Pharmacol 2023; 14:1171293. [PMID: 37274104 PMCID: PMC10232792 DOI: 10.3389/fphar.2023.1171293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/09/2023] [Indexed: 06/06/2023] Open
Abstract
The increasing application of nuclear technology, the high fatality of acute radiation syndrome (ARS) and its complex mechanism make ARS a global difficulty that requires urgent attention. Here we reported that the death receptor 5 (DR5), as well as its ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), were both significantly upregulated after irradiation in mice with 6 Gy γ-ray single radiation. And by intravenously administrated with soluble DR5 fusion protein (sDR5-Fc), the competitive antagonist of DR5, the excessive apoptosis in the radiation-sensitive tissues such as spleen and thymus were significantly inhibited and the radiation-induced damage of spleen and thymus were mitigated, while the expression of apoptosis-inhibiting proteins such as Bcl-2 was also significantly upregulated. The biochemical indicators such as serum ALP, AST, ALT, TBIL, K, and Cl levels that affected by radiation, were improved by sDR5-Fc administration. sDR5-Fc can also regulate the number of immune cells and reduce blood cell death. For in vitro studies, it had been found that sDR5-Fc effectively inhibited apoptosis of human small intestinal mucosal epithelial cells and IEC-6 cells using flow cytometry. Finally, survival studies showed that mice administrated with sDR5-Fc after 9 Gy γ-ray single whole body radiation effectively increased the 30-day survival and was in a significant dose-dependent manner. Overall, the findings revealed that DR5/TRAIL-mediated apoptosis pathway had played important roles in the injury of ARS mice, and DR5 probably be a potential target for ARS therapeutics. And the DR5 apoptosis antagonist, sDR5 fusion protein, probably is a promising anti-ARS drug candidate which deserves further investigation.
Collapse
Affiliation(s)
- Danyang Zhao
- School of Life Sciences, Hebei University, Baoding, China
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Lei Yang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Peng Han
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Haihui Zhang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Fanjun Wang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhiyun Meng
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Hui Gan
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhuona Wu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Wenzhong Sun
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Chuan Chen
- School of Life Sciences, Hebei University, Baoding, China
| | - Guifang Dou
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Ruolan Gu
- Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
9
|
Synthesis and Anticancer Evaluation of New Indole-Based Tyrphostin Derivatives and Their ( p-Cymene)dichloridoruthenium(II) Complexes. Int J Mol Sci 2023; 24:ijms24010854. [PMID: 36614289 PMCID: PMC9821196 DOI: 10.3390/ijms24010854] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/24/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
New N-alkylindole-substituted 2-(pyrid-3-yl)-acrylonitriles with putative kinase inhibitory activity and their (p-cymene)Ru(II) piano-stool complexes were prepared and tested for their antiproliferative efficacy in various cancer models. Some of the indole-based derivatives inhibited tumor cell proliferation at (sub-)micromolar concentrations with IC50 values below those of the clinically relevant multikinase inhibitors gefitinib and sorafenib, which served as positive controls. A focus was set on the investigation of drug mechanisms in HCT-116 p53-knockout colon cancer cells in order to evaluate the dependence of the test compounds on p53. Colony formation assays as well as experiments with tumor spheroids confirmed the excellent antineoplastic efficacy of the new derivatives. Their mode of action included an induction of apoptotic caspase-3/7 activity and ROS formation, as well as anti-angiogenic properties. Docking calculations with EGFR and VEGFR-2 identified the two 3-aryl-2-(pyrid-3-yl)acrylonitrile derivatives 2a and 2b as potential kinase inhibitors with a preferential activity against the VEGFR-2 tyrosine kinase. Forthcoming studies will further unveil the underlying mode of action of the promising new derivatives as well as their suitability as an urgently needed novel approach in cancer treatment.
Collapse
|
10
|
Impact of Oxidative Stress on Molecular Mechanisms of Cervical Ripening in Pregnant Women. Int J Mol Sci 2022; 23:ijms232112780. [PMID: 36361572 PMCID: PMC9657514 DOI: 10.3390/ijms232112780] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 11/29/2022] Open
Abstract
Uterine cervix is one of the essential factors in labor and maintaining the proper course of pregnancy. During the last days of gestation, the cervix undergoes extensive changes manifested by transformation from a tight and rigid to one that is soft and able to dilate. These changes can be summarized as “cervical ripening”. Changes in the cervical tissue can be referred to as remodeling of the extracellular matrix. The entire process is the result of a close relationship between biochemical and molecular pathways, which is strictly controlled by inflammatory and endocrine factors. When the production of reactive oxygen species exceeds the antioxidant capacity, oxidative stress occurs. A physiologic increase of reactive oxygen species (ROS) and reactive nitrogen species (RNS) is observed through pregnancy. ROS play important roles as second messengers in many intracellular signaling cascades contributing to the course of gestation. This review considers their involvement in the cervical ripening process, emphasizing the molecular and biochemical pathways and the clinical implications.
Collapse
|
11
|
Obaidi I, Blanco Fernández A, McMorrow T. Curcumin Sensitises Cancerous Kidney Cells to TRAIL Induced Apoptosis via Let-7C Mediated Deregulation of Cell Cycle Proteins and Cellular Metabolism. Int J Mol Sci 2022; 23:ijms23179569. [PMID: 36076967 PMCID: PMC9455736 DOI: 10.3390/ijms23179569] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 12/31/2022] Open
Abstract
Targeted therapies are the most attractive options in the treatment of different tumours, including kidney cancers. Such therapies have entered a golden era due to advancements in research, breakthroughs in scientific knowledge, and a better understanding of cancer therapy mechanisms, which significantly improve the survival rates and life expectancy of patients. The use of tumour necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) as an anticancer therapy has attracted the attention of the scientific community and created great excitement due to its selectivity in targeting cancerous cells with no toxic impacts on normal tissues. However, clinical studies disappointingly showed the emergence of resistance against TRAIL. This study aimed to employ curcumin to sensitise TRAIL-resistant kidney cancerous ACHN cells, as well as to gain insight into the molecular mechanisms of TRAIL sensitization. Curcumin deregulated the expression of apoptosis-regulating micro Ribonucleic Acid (miRNAs), most notably, let-7C. Transfecting ACHN cells with a let-7C antagomir significantly increased the expression of several cell cycle protein, namely beta (β)-catenin, cyclin dependent kinase (CDK)1/2/4/6 and cyclin B/D. Further, it overexpressed the expression of the two key glycolysis regulating proteins including hypoxia-inducible factor 1-alpha (HIF-1α) and pyruvate dehydrogenase kinase 1 (PDK1). Curcumin also suppressed the expression of the overexpressed proteins when added to the antagomir transfected cells. Overall, curcumin targeted ACHN cell cycle and cellular metabolism by promoting the differential expression of let-7C. To the best of our knowledge, this is the first study to mechanistically report the cancer chemosensitisation potential of curcumin in kidney cancer cells via induction of let-7C.
Collapse
Affiliation(s)
- Ismael Obaidi
- NatPro Centre for Natural Product Research, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, D02 W272 Dublin, Ireland
- College of Pharmacy, University of Babylon, Babylon 51002, Iraq
- Correspondence: (I.O.); (T.M.); Tel.: +353-8-6064-2626 (I.O.); +353-1-716-2317 (ext. 6819) (T.M.)
| | - Alfonso Blanco Fernández
- Flow Cytometry Core Technology, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Tara McMorrow
- Centre for Toxicology, School of Biomedical and Biomolecular Sciences, Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
- Correspondence: (I.O.); (T.M.); Tel.: +353-8-6064-2626 (I.O.); +353-1-716-2317 (ext. 6819) (T.M.)
| |
Collapse
|
12
|
Xie B, Yang J, Zhang J. Chrysin sensitizes osteosarcoma cells against TRAIL-induced apoptosis. Cell Biol Int 2022; 46:1825-1833. [PMID: 35979647 DOI: 10.1002/cbin.11879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 05/30/2022] [Accepted: 07/05/2022] [Indexed: 11/10/2022]
Abstract
Identifying novel curative and preventive approaches that can specifically target the osteosarcoma cells (OS) without affecting the normal cells is appreciable. The aim of this study is to investigate the combined effect of chrysin as an apigenin analog with high therapeutic potential and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) on the treatment of Saos-2 and MG-63 cells. Cell viability were determined using MTT method. The rate of apoptosis was assessed by enzyme-linked immunosorbent assay (ELISA) cell death assay and caspase 8 activity assays. The messenger RNA (mRNA) and protein evaluation of candidate genes include Bcl-2, XIAP, c-IAP1, c-IAP2, and c-FLIP were accomplished before and after the treatment by quantitative real-time polymerase chain reaction (PCR) and Western blot analysis, respectively. Our results showed that chrysin synergistically increased the cytotoxic effects of TRAIL as follows: Chrysin plus TRAIL > TRAIL > Chrysin. Chrysin could sensitize both cells against the TRAIL-induced apoptosis, amplify the caspase 8 activity and this outcome is achieved by decreasing the expression levels of antiapoptotic genes. Our findings suggest that Chrysin can sensitize the OS cell lines against TRAIL through induction of the death receptor pathway. Moreover, the combinational therapy of these agents might be the promising therapeutic regimen for improving the clinical efficacy of TRAIL-induced apoptosis in patients with OS.
Collapse
Affiliation(s)
- Bin Xie
- Department of Orthopedics, Yan'an Peoples's Hospital, Yan'an, China
| | - JunQi Yang
- Department of Orthopaedics, Baoji Central Hospital, Baoji, China
| | - Jun Zhang
- Department of Orthopaedics, Baoji Central Hospital, Baoji, China
| |
Collapse
|
13
|
Very N, El Yazidi-Belkoura I. Targeting O-GlcNAcylation to overcome resistance to anti-cancer therapies. Front Oncol 2022; 12:960312. [PMID: 36059648 PMCID: PMC9428582 DOI: 10.3389/fonc.2022.960312] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/19/2022] [Indexed: 12/14/2022] Open
Abstract
In cancer cells, metabolic reprogramming is associated with an alteration of the O-GlcNAcylation homeostasis. This post-translational modification (PTM) that attaches O-GlcNAc moiety to intracellular proteins is dynamically and finely regulated by the O-GlcNAc Transferase (OGT) and the O-GlcNAcase (OGA). It is now established that O-GlcNAcylation participates in many features of cancer cells including a high rate of cell growth, invasion, and metastasis but little is known about its impact on the response to therapies. The purpose of this review is to highlight the role of O-GlcNAc protein modification in cancer resistance to therapies. We summarize the current knowledge about the crosstalk between O-GlcNAcylation and molecular mechanisms underlying tumor sensitivity/resistance to targeted therapies, chemotherapies, immunotherapy, and radiotherapy. We also discuss potential benefits and strategies of targeting O-GlcNAcylation to overcome cancer resistance.
Collapse
Affiliation(s)
- Ninon Very
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Ikram El Yazidi-Belkoura
- Université de Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
- *Correspondence: Ikram El Yazidi-Belkoura,
| |
Collapse
|
14
|
Xing N, Meng X, Wang S. Isobavachalcone: A comprehensive review of its plant sources, pharmacokinetics, toxicity, pharmacological activities and related molecular mechanisms. Phytother Res 2022; 36:3120-3142. [PMID: 35684981 DOI: 10.1002/ptr.7520] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/10/2022] [Accepted: 05/24/2022] [Indexed: 12/14/2022]
Abstract
Isobavachalcone (IBC), also known as isobapsoralcone, is a natural flavonoid widely derived from many medicinal plants, including Fabaceae, Moraceae, and so forth. IBC has been paid more and more attention by researchers in recent years due to its pharmacological activity in many diseases. This review aims to describe in detail the plant sources, pharmacokinetics, toxicity, pharmacological activities, and molecular mechanisms of IBC on various diseases. We found that IBC can be obtained not only by extraction but also by chemical synthesis. Pharmacokinetic studies have shown that IBC has low bioavailability, but can penetrate the blood-brain barrier and is widely distributed in the brain. Its pharmacological activities mainly include anticancer, antibacterial, anti-inflammatory, antiviral, neuroprotective, bone protection, and other activities. In particular, IBC shows strong anti-tumor and anti-inflammatory therapeutic potential due to its anti-cancer and anti-inflammatory activities. However, due to its hepatotoxicity, there may be more drug interactions. Therefore, more and more in-depth studies are needed for its clinical application. Mechanically, IBC can induce the production of reactive oxygen species (ROS), inhibit AKT, ERK, and Wnt pathways, and promote apoptosis of cancer cells through mitochondrial or endoplasmic reticulum pathways. IBC can inhibit the NF-κB pathway and the production of multiple inflammatory mediators by activating NRF2/HO-1 pathway, thus producing anti-inflammatory effects. Moreover, we discussed the limitations of current research on IBC and put forward some new perspectives and challenges, which provide a strong basis for clinical application and new drug development of IBC in the future.
Collapse
Affiliation(s)
- Nan Xing
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shaohui Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
Feng X, Li F, Zhang L, Liu W, Wang X, Zhu R, Qiao ZA, Yu B, Yu X. TRAIL-modified, doxorubicin-embedded periodic mesoporous organosilica nanoparticles for targeted drug delivery and efficient antitumor immunotherapy. Acta Biomater 2022; 143:392-405. [PMID: 35259519 DOI: 10.1016/j.actbio.2022.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 12/17/2022]
Abstract
Traditional anticancer treatments directly target tumor cells. In contrast, cancer immunotherapy fortifies host immunity. Nanoparticles that incorporate both immunomodulatory and chemotherapeutic agents regulate the tumor microenvironment by activating immune cells and enhancing antitumor immunity. Nanoparticle-based cancer immunotherapy has received considerable attention and has been extensively studied in recent years. In this study, we developed a targeted drug delivery system to enhance immunotherapeutic efficacy and overcome drug resistance by inducing tumor apoptosis and immunogenic cell death (ICD), and activating immune cells. Periodic mesoporous organosilica nanoparticles (PMOs) bore tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) on their surfaces, and their inner cores were loaded with doxorubicin (DOX). TRAIL enhanced the nanoparticle-targeting capacity and worked synergistically with DOX against breast cancer cells in vitro and in vivo. Furthermore, we revealed for the first time the ability of PMOs to activate dendritic cells (DCs) and elevate ICD levels of DOX in vitro, and TRAIL further enhances the immunomodulatory function of PMOs. Systemic exposure to DOX@PMO-hT induced an immune response, activated DCs and CD4+ and CD8+ T cells, and significantly suppressed tumor growth in a 4T1-bearing immunocompetent mouse model. Overall, our study demonstrates that TRAIL-modified, DOX-embedded PMO nanoparticles represent a good candidate for tumor-targeted immunotherapy, which has relatively superior therapeutic efficacy and highly promising future application prospects. STATEMENT OF SIGNIFICANCE: This study revealed for the first time the ability of PMOs to elevate ICD levels and activate DCs in vitro. The results explained the immunomodulatory function of PMOs and demonstrated the synergistic effects of TRAIL and DOX in triple-negative breast cancer. In addition, immunomodulatory effects of the drug delivery vectors constructed in this study were verified in vivo.
Collapse
|
16
|
Rana D, Salave S, Perla A, Nadkarni A, Kohle S, Jindal AB, Mandoli A, Dwivedi P, Benival D. Bugs as Drugs: Understanding the Linkage between Gut Microbiota and Cancer Treatment Microbiome in Cancer Therapy. Curr Drug Targets 2022; 23:869-888. [PMID: 35264088 DOI: 10.2174/1389450123666220309101345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/03/2022] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND The commensal microbiota is known to regulate host physiology. Dysbiosis or compromised Resilience in the microbial ecology is related to the impending risk of cancer. A potential link between cancer and microbiota is indicated by a lot of evidence. OBJECTIVE The current review explores in detail the various links leading to and /or facilitating oncogenesis, providing sound reasoning or a basis for its utilization as potential therapeutic targets. The present review emphasizes the existing knowledge of the microbiome in cancer and further elaborates on the factors like genetic modifications, effects of dietary components, and environmental agents that are considered to assess the direct and indirect effect of microbes in the process of oncogenesis and on the host's health. Strategies modulating the microbiome and novel biotherapeutics are also discussed. Pharmacomicrobiomics is one such niche accounting for the interplay between the microbiome, xenobiotic, and host responses is also looked upon. METHODS The literature search strategy for this review was conducted by following the methodology of the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA). The method includes the collection of data from different search engines like PubMed, ScienceDirect, SciFinder etc. to get coverage of relevant literature for accumulating appropriate information regarding microbiome, cancer, and their linkages. RESULTS These considerations are made to expand the existing literature on the role of gut microbiota on the host's health, the interaction between host and microbiota, and the reciprocal relationship between the microbiome and modified neoplastic cells. CONCLUSION Potential therapeutic implications of cancer microbiomes that are yet unexplored and have rich therapeutic dividends improving human health are discussed in detail in this review.
Collapse
Affiliation(s)
- Dhwani Rana
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), 382355, India
| | - Sagar Salave
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), 382355, India
| | - Akhil Perla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), 382355, India
| | - Akanksha Nadkarni
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), 382355, India
| | - Shital Kohle
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), 382355, India
| | - Anil B Jindal
- Department of Pharmacy, Birla Institute of Technology and Science Pilani (BITS PILANI), Pilani Campus, Rajasthan, 333031, India
| | - Amit Mandoli
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), 382355, India
| | - Pradeep Dwivedi
- Department of Pharmacology, All India Institute of Medical Sciences- Jodhpur (AIIMS), 342005, India
| | - Derajram Benival
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), 382355, India
| |
Collapse
|
17
|
Behind the Adaptive and Resistance Mechanisms of Cancer Stem Cells to TRAIL. Pharmaceutics 2021; 13:pharmaceutics13071062. [PMID: 34371753 PMCID: PMC8309156 DOI: 10.3390/pharmaceutics13071062] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 12/20/2022] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), also known as Apo-2 ligand (Apo2L), is a member of the TNF cytokine superfamily. TRAIL has been widely studied as a novel strategy for tumor elimination, as cancer cells overexpress TRAIL death receptors, inducing apoptosis and inhibiting blood vessel formation. However, cancer stem cells (CSCs), which are the main culprits responsible for therapy resistance and cancer remission, can easily develop evasion mechanisms for TRAIL apoptosis. By further modifying their properties, they take advantage of this molecule to improve survival and angiogenesis. The molecular mechanisms that CSCs use for TRAIL resistance and angiogenesis development are not well elucidated. Recent research has shown that proteins and transcription factors from the cell cycle, survival, and invasion pathways are involved. This review summarizes the main mechanism of cell adaption by TRAIL to promote response angiogenic or pro-angiogenic intermediates that facilitate TRAIL resistance regulation and cancer progression by CSCs and novel strategies to induce apoptosis.
Collapse
|
18
|
Quiroz-Reyes AG, Delgado-Gonzalez P, Islas JF, Gallegos JLD, Martínez Garza JH, Garza-Treviño EN. Behind the Adaptive and Resistance Mechanisms of Cancer Stem Cells to TRAIL. Pharmaceutics 2021; 13:1062. [DOI: https:/doi.org/10.3390/pharmaceutics13071062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), also known as Apo-2 ligand (Apo2L), is a member of the TNF cytokine superfamily. TRAIL has been widely studied as a novel strategy for tumor elimination, as cancer cells overexpress TRAIL death receptors, inducing apoptosis and inhibiting blood vessel formation. However, cancer stem cells (CSCs), which are the main culprits responsible for therapy resistance and cancer remission, can easily develop evasion mechanisms for TRAIL apoptosis. By further modifying their properties, they take advantage of this molecule to improve survival and angiogenesis. The molecular mechanisms that CSCs use for TRAIL resistance and angiogenesis development are not well elucidated. Recent research has shown that proteins and transcription factors from the cell cycle, survival, and invasion pathways are involved. This review summarizes the main mechanism of cell adaption by TRAIL to promote response angiogenic or pro-angiogenic intermediates that facilitate TRAIL resistance regulation and cancer progression by CSCs and novel strategies to induce apoptosis.
Collapse
|
19
|
Bomba HN, Sheets KT, Valdivia A, Khagi S, Ruterbories L, Mariani CL, Borst LB, Tokarz DA, Hingtgen SD. Personalized-induced neural stem cell therapy: Generation, transplant, and safety in a large animal model. Bioeng Transl Med 2021; 6:e10171. [PMID: 33532581 PMCID: PMC7823134 DOI: 10.1002/btm2.10171] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/28/2020] [Accepted: 06/29/2020] [Indexed: 12/24/2022] Open
Abstract
In this study, we take an important step toward clinical translation by generating the first canine-induced neural stem cells (iNSCs). We explore key aspects of scale-up, persistence, and safety of personalized iNSC therapy in autologous canine surgery models. iNSCs are a promising new approach to treat aggressive cancers of the brain, including the deadly glioblastoma. Created by direct transdifferentiation of fibroblasts, iNSCs are known to migrate through the brain, track down invasive cancer foci, and deliver anticancer payloads that significantly reduce tumor burden and extend survival of tumor-bearing mice. Here, skin biopsies were collected from canines and converted into the first personalized canine iNSCs engineered to carry TNFα-related apoptosis-inducing ligand (TRAIL) and thymidine kinase (TK), as well as magnetic resonance imaging (MRI) contrast agents for in vivo tracking. Time-lapse analysis showed canine iNSCs efficiently migrate to human tumor cells, and cell viability assays showed both TRAIL and TK monotherapy markedly reduced tumor growth. Using intraoperative navigation and two delivery methods to closely mimic human therapy, canines received autologous iNSCs either within postsurgical cavities in a biocompatible matrix or via a catheter placed in the lateral ventricle. Both strategies were well tolerated, and serial MRI showed hypointense regions at the implant sites that remained stable through 86 days postimplant. Serial fluid sample testing following iNSC delivery showed the bimodal personalized therapy was well tolerated, with no iNSC-induced abnormal tissue pathology. Overall, this study lays an important foundation as this promising personalized cell therapy advances toward human patient testing.
Collapse
Affiliation(s)
- Hunter N. Bomba
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Kevin T. Sheets
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Alain Valdivia
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Simon Khagi
- Department of NeurosurgeryThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Laura Ruterbories
- Department of Clinical Sciences, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Christopher L. Mariani
- Department of Clinical Sciences, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Luke B. Borst
- Department of Population Health and Pathobiology, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Debra A. Tokarz
- Department of Population Health and Pathobiology, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Shawn D. Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
20
|
Boi SK, Orlandella RM, Gibson JT, Turbitt WJ, Wald G, Thomas L, Buchta Rosean C, Norris KE, Bing M, Bertrand L, Gross BP, Makkouk A, Starenki D, Farag KI, Sorge RE, Brown JA, Gordetsky J, Yasin H, Garje R, Nandagopal L, Weiner GJ, Lubaroff DM, Arend RC, Li P, Zakharia Y, Yang E, Salem AK, Nepple K, Marquez-Lago TT, Norian LA. Obesity diminishes response to PD-1-based immunotherapies in renal cancer. J Immunother Cancer 2020; 8:e000725. [PMID: 33427691 PMCID: PMC7757487 DOI: 10.1136/jitc-2020-000725] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Obesity is a major risk factor for renal cancer, yet our understanding of its effects on antitumor immunity and immunotherapy outcomes remains incomplete. Deciphering these associations is critical, given the growing clinical use of immune checkpoint inhibitors for metastatic disease and mounting evidence for an obesity paradox in the context of cancer immunotherapies, wherein obese patients with cancer have improved outcomes. METHODS We investigated associations between host obesity and anti-programmed cell death (PD-1)-based outcomes in both renal cell carcinoma (RCC) subjects and orthotopic murine renal tumors. Overall survival (OS) and progression-free survival (PFS) were determined for advanced RCC subjects receiving standard of care anti-PD-1 who had ≥6 months of follow-up from treatment initiation (n=73). Renal tumor tissues were collected from treatment-naive subjects categorized as obese (body mass index, 'BMI' ≥30 kg/m2) or non-obese (BMI <30 kg/m2) undergoing partial or full nephrectomy (n=19) then used to evaluate the frequency and phenotype of intratumoral CD8+ T cells, including PD-1 status, by flow cytometry. In mice, antitumor immunity and excised renal tumor weights were evaluated ±administration of a combinatorial anti-PD-1 therapy. For a subset of murine renal tumors, immunophenotyping was performed by flow cytometry and immunogenetic profiles were evaluated via nanoString. RESULTS With obesity, RCC patients receiving anti-PD-1 administration exhibited shorter PFS (p=0.0448) and OS (p=0.0288). Treatment-naive renal cancer subjects had decreased frequencies of tumor-infiltrating PD-1highCD8+ T cells, a finding recapitulated in our murine model. Following anti-PD-1-based immunotherapy, both lean and obese mice possessed distinct populations of treatment responders versus non-responders; however, obesity reduced the frequency of treatment responders (73% lean vs 44% obese). Tumors from lean and obese treatment responders displayed similar immunogenetic profiles, robust infiltration by PD-1int interferon (IFN)γ+CD8+ T cells and reduced myeloid-derived suppressor cells (MDSC), yielding favorable CD44+CD8+ T cell to MDSC ratios. Neutralizing interleukin (IL)-1β in obese mice improved treatment response rates to 58% and reduced MDSC accumulation in tumors. CONCLUSIONS We find that obesity is associated with diminished efficacy of anti-PD-1-based therapies in renal cancer, due in part to increased inflammatory IL-1β levels, highlighting the need for continued study of this critical issue.
Collapse
Affiliation(s)
- Shannon K Boi
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rachael M Orlandella
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Justin Tyler Gibson
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - William James Turbitt
- Department of Nutrition Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Gal Wald
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Lewis Thomas
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Claire Buchta Rosean
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Katlyn E Norris
- Honors Undergraduate Research Program, School of Health Professions, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Megan Bing
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Laura Bertrand
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Brett P Gross
- Interdisciplinary Program in Immunology, The University of Iowa, Iowa City, Iowa, USA
| | - Amani Makkouk
- Interdisciplinary Program in Immunology, The University of Iowa, Iowa City, Iowa, USA
| | - Dmytro Starenki
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Kristine I Farag
- Science and Technology Honors Program, College of Arts and Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Robert E Sorge
- Department of Psychology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James A Brown
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
| | - Jennifer Gordetsky
- Departments of Pathology and Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hesham Yasin
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
- Department of Internal Medicine, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Rohan Garje
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
- Genitourinary Oncology Program, Division of Hematology, Oncology and Blood and Marrow Transplantation, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Lakshminarayanan Nandagopal
- Division of Hematology and Oncology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
- The University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama, USA
| | - George J Weiner
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
- Department of Internal Medicine, Division of General Medicine, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - David M Lubaroff
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
| | - Rebecca C Arend
- The University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama, USA
- Department of Obstetrics and Gynecology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Peng Li
- Department of Biostatistics, The University of Alabama at Birmingham School of Nursing, Birmingham, Alabama, USA
| | - Yousef Zakharia
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
- Genitourinary Oncology Program, Division of Hematology, Oncology and Blood and Marrow Transplantation, The University of Iowa, Iowa City, Iowa, USA
| | - Eddy Yang
- The University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama, USA
- Department of Radiation Oncology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Aliasger K Salem
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
- Division of Pharmaceutics and Translational Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa, USA
| | - Kenneth Nepple
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
| | - Tatiana T Marquez-Lago
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lyse A Norian
- Department of Nutrition Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- The University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama, USA
| |
Collapse
|
21
|
Gao S, Fang Y, Tu S, Chen H, Shao A. Insight into the divergent role of TRAIL in non-neoplastic neurological diseases. J Cell Mol Med 2020; 24:11070-11083. [PMID: 32827246 PMCID: PMC7576257 DOI: 10.1111/jcmm.15757] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/04/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
Tumour necrosis factor–related apoptosis‐inducing ligand (TRAIL) is a member of the tumour necrosis factor (TNF) superfamily which mainly induces apoptosis of tumour cells and transformed cell lines with no systemic toxicity, whereas they share high sequence homology with TNF and CD95L. These unique effects of TRAIL have made it an important molecule in oncology research. However, the research on TRAIL‐related antineoplastic agents has lagged behind and has been limited by the extensive drug resistance in cancer cells. Given the several findings showing that TRAIL is involved in immune regulation and other pleiotropic biological effects in non‐malignant cells, TRAIL and its receptors have attracted widespread attention from researchers. In the central nervous system (CNS), TRAIL is highly correlated with malignant tumours such as glioma and other non‐neoplastic disorders such as acute brain injury, CNS infection and neurodegenerative disease. Many clinical and animal studies have revealed the dual roles of TRAIL in which it causes damage by inducing cell apoptosis, and confers protection by enhancing both pro‐ and non‐apoptosis effects in different neurological disorders and at different sites or stages. Its pro‐apoptotic effect produces a pro‐survival effect that cannot be underestimated. This review extensively covers in vitro and in vivo experiments and clinical studies investigating TRAIL. It also provides a summary of the current knowledge on the TRAIL signalling pathway and its involvement in pathogenesis, diagnosis and therapeutics of CNS disorders as a basis for future research.
Collapse
Affiliation(s)
- Shiqi Gao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanjian Fang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Tu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Huaijun Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
22
|
Kurmi BD, Patel P, Paliwal R, Paliwal SR. Molecular approaches for targeted drug delivery towards cancer: A concise review with respect to nanotechnology. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101682] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
23
|
Regulation of pancreatic cancer TRAIL resistance by protein O-GlcNAcylation. J Transl Med 2020; 100:777-785. [PMID: 31896813 PMCID: PMC7183418 DOI: 10.1038/s41374-019-0365-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/26/2019] [Accepted: 12/07/2019] [Indexed: 12/11/2022] Open
Abstract
TRAIL-activating therapy is promising in treating various cancers, including pancreatic cancer, a highly malignant neoplasm with poor prognosis. However, many pancreatic cancer cells are resistant to TRAIL-induced apoptosis despite their expression of intact death receptors (DRs). Protein O-GlcNAcylation is a versatile posttranslational modification that regulates various biological processes. Elevated protein O-GlcNAcylation has been recently linked to cancer cell growth and survival. In this study, we evaluated the role of protein O-GlcNAcylation in pancreatic cancer TRAIL resistance, and identified higher levels of O-GlcNAcylation in TRAIL-resistant pancreatic cancer cells. With gain- and loss-of-function of the O-GlcNAc-adding enzyme, O-GlcNActransferase (OGT), we determined that increasing O-GlcNAcylation rendered TRAIL-sensitive cells more resistant to TRA-8-induced apoptosis, while inhibiting O-GlcNAcylation promoted TRA-8-induced apoptosis in TRAIL-resistance cells. Furthermore, we demonstrated that OGT knockdown sensitized TRAIL-resistant cells to TRA-8 therapy in a mouse model in vivo. Mechanistic studies revealed direct O-GlcNAc modifications of DR5, which regulated TRA-8-induced DR5 oligomerization. We further defined that DR5 O-GlcNAcylation was independent of FADD, the adapter protein for the downstream death-inducing signaling. These studies have demonstrated an important role of protein O-GlcNAcylation in regulating TRAIL resistance of pancreatic cancer cells; and uncovered the contribution of O-GlcNAcylation to DR5 oligomerization and thus mediating DR-inducing signaling.
Collapse
|
24
|
Interactions of Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) with the Immune System: Implications for Inflammation and Cancer. Cancers (Basel) 2019; 11:cancers11081161. [PMID: 31412671 PMCID: PMC6721490 DOI: 10.3390/cancers11081161] [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: 06/30/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/24/2022] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily. TRAIL has historically been distinct from the Fas ligand and TNFα in terms of selective apoptosis induction in tumor cells and has a nearly non-existent systemic toxicity. Consequently, in the search for an ideal drug for tumor therapy, TRAIL rapidly drew interest, promising effective tumor control with minimal side effects. However, euphoria gave way to disillusionment as it turned out that carcinoma cells possess or can acquire resistance to TRAIL-induced apoptosis. Additionally, studies on models of inflammation and autoimmunity revealed that TRAIL can influence immune cells in many different ways. While TRAIL was initially found to be an important player in tumor defense by natural killer cells or cytotoxic T cells, additional effects of TRAIL on regulatory T cells and effector T cells, as well as on neutrophilic granulocytes and antigen-presenting cells, became focuses of interest. The tumor-promoting effects of these interactions become particularly important for consideration in cases where tumors are resistant to TRAIL-induced apoptosis. Consequently, murine models have shown that TRAIL can impair the tumor microenvironment toward a more immunosuppressive type, thereby promoting tumor growth. This review summarizes the current state of knowledge on TRAIL’s interactions with the immune system in the context of cancer.
Collapse
|
25
|
Song T, Wang P, Yu X, Wang A, Chai G, Fan Y, Zhang Z. Systems analysis of phosphorylation-regulated Bcl-2 interactions establishes a model to reconcile the controversy over the significance of Bcl-2 phosphorylation. Br J Pharmacol 2019; 176:491-504. [PMID: 30500985 PMCID: PMC6329625 DOI: 10.1111/bph.14555] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 10/23/2018] [Accepted: 10/25/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE The biological significance of the multi-site phosphorylation of Bcl-2 at its loop region (T69, S70 and S87) has remained controversial for decades. This is a major obstacle for understanding apoptosis and anti-tumour drug development. EXPERIMENTAL APPROACH We established a mathematical model into which a phosphorylation and de-phosphorylation process of Bcl-2 was integrated. Paclitaxel-treated breast cancer cells were used as experimental models. Changes in the kinetics of binding with its critical partners, induced by phosphorylation of Bcl-2 were experimentally obtained by surface plasmon resonance, using a phosphorylation-mimicking mutant EEE-Bcl-2 (T69E, S70E and S87E). KEY RESULTS Mathematical simulations combined with experimental validation showed that phosphorylation regulates Bcl-2 with different dynamics depending on the extent of Bcl-2 phosphorylation and the phosphorylated Bcl-2-induced changes in binding kinetics. In response to Bcl-2 homology 3 (BH3)-only protein Bmf stress, Bcl-2 phosphorylation switched from diminishing to enhancing the Bcl-2 anti-apoptotic ability with increased phosphorylation of Bcl-2, and the turning point was 50% Bcl-2 phosphorylation induced by 0.2 μM paclitaxel treatment. In contrast, Bcl-2 phosphorylation enhanced the anti-apoptotic ability of Bcl-2 towards other BH3-only proteins Bim, Bad and Puma, throughout the entire phosphorylation procedure. CONCLUSIONS AND IMPLICATIONS The model could accurately predict the effects of anti-tumour drugs that involve the Bcl-2 family pathway, as shown with ABT-199 or etoposide.
Collapse
Affiliation(s)
- Ting Song
- State Key Laboratory of Fine Chemicals, School of ChemistryDalian University of TechnologyDalianChina
| | - Peiran Wang
- State Key Laboratory of Fine Chemicals, School of ChemistryDalian University of TechnologyDalianChina
| | - Xiaoyan Yu
- School of Life Science and TechnologyDalian University of TechnologyDalianChina
| | - Anhui Wang
- School of Innovation ExperimentDalian University of TechnologyDalianChina
| | - Gaobo Chai
- School of Life Science and TechnologyDalian University of TechnologyDalianChina
| | - Yudan Fan
- School of Life Science and TechnologyDalian University of TechnologyDalianChina
| | - Zhichao Zhang
- State Key Laboratory of Fine Chemicals, School of ChemistryDalian University of TechnologyDalianChina
| |
Collapse
|
26
|
Xu F, Sun Y, Yang SZ, Zhou T, Jhala N, McDonald J, Chen Y. Cytoplasmic PARP-1 promotes pancreatic cancer tumorigenesis and resistance. Int J Cancer 2019; 145:474-483. [PMID: 30614530 DOI: 10.1002/ijc.32108] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/26/2018] [Accepted: 12/20/2018] [Indexed: 12/16/2022]
Abstract
The poly(ADP-ribose) polymerases (PARP) play important roles in repairing damaged DNA during intrinsic cell death. We recently linked PARP-1 to death receptor (DR)-activated extrinsic apoptosis, the present studies sought to elucidate the function of cytoplasmic PARP-1 in pancreatic cancer tumorigenesis and therapy. Using human normal and pancreatic cancer tissues, we analyzed the prevalence of cytoplasmic PARP-1 expression. In normal human pancreatic tissues, PARP-1 expression was present in the nucleus; however, cytoplasmic PARP-1 expression was identified in pancreatic cancers. Therefore, cytoplasmic PARP-1 mutants were generated by site-direct mutagenesis, to determine a causative effect of cytoplasmic PARP-1 on pancreatic cancer tumorigenesis and sensitivity to therapy with TRA-8, a humanized DR5 antibody. PARP-1 cytoplasmic mutants rendered TRA-8 sensitive pancreatic cancer cells, BxPc-3 and MiaPaCa-2, more resistant to TRA-8-induced apoptosis; whereas wild-type PARP-1, localizing mainly in the nucleus, had no effects. Additionally, cytoplasmic PARP-1, but not wild-type PARP-1, increased resistance of BxPc-3 cells to TRA-8 therapy in a mouse xenograft model in vivo. Inhibition of PARP enzymatic activity attenuated cytoplasmic PARP-1-mediated TRA-8 resistance. Furthermore, increased cytoplasmic PARP-1, but not wild-type PARP-1, was recruited into the TRA-8-activated death-inducing signaling complex and associated with increased and sustained activation of Src-mediated survival signals. In contrast, PARP-1 knockdown inhibited Src activation. Taken together, we have identified a novel function and mechanism underlying cytoplasmic PARP-1, distinct from nuclear PARP-1, in regulating DR5-activated apoptosis. Our studies support an innovative application of available PARP inhibitors or new cytoplasmic PARP-1 antagonists to enhance TRAIL therapy for TRAIL-resistant pancreatic cancers.
Collapse
Affiliation(s)
- Fei Xu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL.,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Sun
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Shan-Zhong Yang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Tong Zhou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Nirag Jhala
- Department of Pathology, Temple University, Philadelphia, PA
| | - Jay McDonald
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL.,Birmingham Veterans Affairs Medical Center, Research Department, Birmingham, AL
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL.,Birmingham Veterans Affairs Medical Center, Research Department, Birmingham, AL
| |
Collapse
|
27
|
Haldar R, Shaashua L, Lavon H, Lyons YA, Zmora O, Sharon E, Birnbaum Y, Allweis T, Sood AK, Barshack I, Cole S, Ben-Eliyahu S. Perioperative inhibition of β-adrenergic and COX2 signaling in a clinical trial in breast cancer patients improves tumor Ki-67 expression, serum cytokine levels, and PBMCs transcriptome. Brain Behav Immun 2018; 73:294-309. [PMID: 29800703 DOI: 10.1016/j.bbi.2018.05.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/10/2018] [Accepted: 05/18/2018] [Indexed: 12/18/2022] Open
Abstract
Catecholamines and prostaglandins are secreted abundantly during the perioperative period in response to stress and surgery, and were shown by translational studies to promote tumor metastasis. Here, in a phase-II biomarker clinical trial in breast cancer patients (n = 38), we tested the combined perioperative use of the β-blocker, propranolol, and the COX2-inhibitor, etodolac, scheduled for 11 consecutive perioperative days, starting 5 days before surgery. Blood samples were taken before treatment (T1), on the mornings before and after surgery (T2&T3), and after treatment cessation (T4). Drugs were well tolerated. Results based on a-priori hypotheses indicated that already before surgery (T2), serum levels of pro-inflammatory IL-6, CRP, and IFNγ, and anti-inflammatory, cortisol and IL-10, increased. At T2 and/or T3, drug treatment reduced serum levels of the above pro-inflammatory cytokines and of TRAIL, as well as activity of multiple inflammation-related transcription factors (including NFκB, STAT3, ISRE), but not serum levels of cortisol, IL-10, IL-18, IL-8, VEGF and TNFα. In the excised tumor, treatment reduced the expression of the proliferation marker Ki-67, and positively affected its transcription factors SP1 and AhR. Exploratory analyses of transcriptome modulation in PBMCs revealed treatment-induced improvement at T2/T3 in several transcription factors that in primary tumors indicate poor prognosis (CUX1, THRa, EVI1, RORa, PBX1, and T3R), angiogenesis (YY1), EMT (GATA1 and deltaEF1/ZEB1), proliferation (GATA2), and glucocorticoids response (GRE), while increasing the activity of the oncogenes c-MYB and N-MYC. Overall, the drug treatment may benefit breast cancer patients through reducing systemic inflammation and pro-metastatic/pro-growth biomarkers in the excised tumor and PBMCs.
Collapse
Affiliation(s)
- Rita Haldar
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel
| | - Lee Shaashua
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel
| | - Hagar Lavon
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel
| | - Yasmin A Lyons
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, M.D. Anderson Cancer Center at University of Texas, Huston, TX, USA
| | - Oded Zmora
- Department of Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
| | - Eran Sharon
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petach-Tikva, Israel
| | - Yehudit Birnbaum
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petach-Tikva, Israel
| | - Tanir Allweis
- Department of Surgery, Kaplan Medical Center, Rehovot, Israel
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, M.D. Anderson Cancer Center at University of Texas, Huston, TX, USA
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Ramat Gan, Israel
| | - Steve Cole
- Department of Medicine, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Shamgar Ben-Eliyahu
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel.
| |
Collapse
|
28
|
Lu Z, Zhang G, Zhang Y, Hua P, Fang M, Wu M, Liu T. Isoalantolactone induces apoptosis through reactive oxygen species-dependent upregulation of death receptor 5 in human esophageal cancer cells. Toxicol Appl Pharmacol 2018; 352:46-58. [DOI: 10.1016/j.taap.2018.05.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/15/2018] [Accepted: 05/21/2018] [Indexed: 01/20/2023]
|
29
|
Zhao X, Zhang C, Le Z, Zeng S, Pan C, Shi J, Wang J, Zhao X. Telomerase reverse transcriptase interference synergistically promotes tumor necrosis factor‑related apoptosis‑inducing ligand‑induced oral squamous cell carcinoma apoptosis and suppresses proliferation in vitro and in vivo. Int J Mol Med 2018; 42:1283-1294. [PMID: 29901096 PMCID: PMC6089774 DOI: 10.3892/ijmm.2018.3721] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 05/30/2018] [Indexed: 11/09/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is known to induce cell apoptosis in many types of cancer cells. However, some malignant cells still exhibit anti-apoptosis features induced by TRAIL; thus the underlying mechanisms that regulate sensitivity and resistance of tumor cells to TRAIL-induced apoptosis remain unclear. Human telomerase reverse transcriptase (hTERT) is overexpressed in most types of human tumors and is mostly inactive in somatic cells. The present study aimed to investigate the endogenous effects and mechanisms of hTERT inhibition and TRAIL overexpression on TRAIL-induced apoptosis of human oral squamous cell carcinoma (OSCC) cells. The effects of adeno-associated virus (AAV)-mediated TRAIL and hTERT gene silencing by RNA interference were investigated on the proliferation and apoptosis of human OSCC cells in vitro and in vivo. The present results suggest that knockdown of hTERT expression accelerated TRAIL-resistant OSCC cells to TRAIL-induced apoptosis and impaired OSCC cell proliferation. In addition, this process is accompanied by the upregulation of caspase-3, caspase-8 and caspase-9, and downregulation of B cell lymphoma-2. Additionally, the possible mechanisms underlying the association between TRAIL expression and hTERT silencing were explored. The results demonstrated that TRAIL expression levels were elevated when the hTERT gene was silenced, and notable anti-tumor effects were observed when TRAIL upregulation and hTERT gene silencing were carried out simultaneously. The present findings provide experimental evidence for the combined use of TRAIL and hTERT as a possible gene therapy strategy in oral cancer.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Stomatology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Cuicui Zhang
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| | - Zhiliang Le
- Department of Stomatology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510635, P.R. China
| | - Suyun Zeng
- Department of Periodontology, Hefei Stomatological Hospital, Hefei, Anhui 230001, P.R. China
| | - Chaobin Pan
- Department of Oral and Maxillofacial Surgery, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jianjie Shi
- Department of Stomatology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Jianguang Wang
- Department of Oral and Maxillofacial Surgery, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Xiaopeng Zhao
- Department of Oral and Maxillofacial Surgery, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
30
|
Shi Y, Pang X, Wang J, Liu G. NanoTRAIL-Oncology: A Strategic Approach in Cancer Research and Therapy. Adv Healthc Mater 2018. [PMID: 29527836 DOI: 10.1002/adhm.201800053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
TRAIL is a member of the tumor necrosis factor superfamily that can largely trigger apoptosis in a wide variety of cancer cells, but not in normal cells. However, insufficient exposure to cancer tissues or cells and drug resistance has severely impeded the clinical application of TRAIL. Recently, nanobiotechnology has brought about a revolution in advanced drug delivery for enhanced anticancer therapy using TRAIL. With the help of materials science, immunology, genetic engineering, and protein engineering, substantial progress is made by expressing fusion proteins with TRAIL, engineering TRAIL on biological membranes, and loading TRAIL into functional nanocarriers or conjugating it onto their surfaces. Thus, the nanoparticle-based TRAIL (nanoTRAIL) opens up intriguing opportunities for efficient and safe bioapplications. In this review, the mechanisms of action and biological function of TRAIL, as well as the current status of TRAIL treatment, are comprehensively discussed. The application of functional nanotechnology combined with TRAIL in cancer therapy is also discussed.
Collapse
Affiliation(s)
- Yesi Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine; School of Public Health; Xiamen University; Xiamen 361102 China
| | - Xin Pang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine; School of Public Health; Xiamen University; Xiamen 361102 China
| | - Junqing Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine; School of Public Health; Xiamen University; Xiamen 361102 China
- Collaborative Innovation Center of Guangxi Biological Medicine and the; Medical and Scientific Research Center; Guangxi Medical University; Nanning 530021 China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine; School of Public Health; Xiamen University; Xiamen 361102 China
| |
Collapse
|
31
|
Takahashi T, Mine Y, Okamoto T. Intracellular reduction of coenzyme Q homologues with a short isoprenoid side chain induces apoptosis of HeLa cells. J Biochem 2018; 163:329-339. [PMID: 29319808 DOI: 10.1093/jb/mvy002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/21/2017] [Indexed: 11/13/2022] Open
Abstract
Coenzyme Q (CoQ) is an essential factor of the mitochondrial respiratory chain. CoQ homologues with different lengths of the isoprenoid side chain are widely distributed in nature, but little is known about the relationship between the isoprenoid side chain length and biological function; therefore, we examined the effects of CoQ homologues on HeLa cells. When CoQ homologues with a shorter isoprenoid side chain than CoQ4 were added to HeLa cells, they induced cell death, and the order of cytotoxic intensity was as follows: CoQ0 ≫ CoQ3 ≈ CoQ1 > CoQ2 ≫ CoQ4. Furthermore, we found that CoQ1, CoQ2 and CoQ3 could induce caspase-mediated apoptosis, and the order of intensity was as follows: CoQ3 > CoQ2 ≥ CoQ1. We could not identify the participation of reactive oxygen species in the apoptosis induction, but observed that an NAD(P)H dehydrogenase (quinone) 1 (NQO1) inhibitor, dicumarol, could inhibit not only the intracellular reduction of the homologues but also apoptosis. However, because dicumarol did not affect well-known apoptosis inducers, such as anti-Fas IgG, tumor necrosis factor (TNF)-α, TNF-related apoptosis-inducing ligand, UV-B and H2O2 of HeLa cells at all, we concluded that NQO1-related intracellular reduction of CoQ, or its reduced product, ubiquinol, may participate in the apoptosis induction of HeLa cells.
Collapse
Affiliation(s)
- Takayuki Takahashi
- Laboratory of Biochemistry, Department of Health Science and Social Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan
| | - Yukitoshi Mine
- Laboratory of Biochemistry, Department of Health Science and Social Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan
| | - Tadashi Okamoto
- Laboratory of Biochemistry, Department of Health Science and Social Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan
| |
Collapse
|
32
|
Ebrahimian M, Taghavi S, Ghoreishi M, Sedghi S, Amel Farzad S, Ramezani M, Hashemi M. Evaluation of Efficiency of Modified Polypropylenimine (PPI) with Alkyl Chains as Non-viral Vectors Used in Co-delivery of Doxorubicin and TRAIL Plasmid. AAPS PharmSciTech 2018; 19:1029-1036. [PMID: 29116619 DOI: 10.1208/s12249-017-0913-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/23/2017] [Indexed: 11/30/2022] Open
Abstract
In this study, co-delivery system was achieved via plasmid encoding TNF related apoptosis inducing ligand (pTRAIL) and doxorubicin (DOX) using carrier based on polypropylenimine (PPI) modified with 10-bromodecanoic acid. Incorporation of alkylcarboxylate chain to PPIs (G4 and G5) could improve transfection efficiency via overcoming the plasma membrane barrier of the cells and decrease cytotoxicity of PPI. Characterization of fabricated NPs revealed that PPI G5 in which 30% of primary amines were substituted by alkyl carboxylate chain (PPI G5-Alkyl 30%) has higher drug loading as compared to the other formulations. PPI G5-Alkyl 30% indicated a decreased drug release may be due to alkyl chains on the surface of PPI, which serve as an additional hindrance for drug diffusion. In vitro cytotoxicity experiments demonstrated that co-delivery system induced apoptosis of tumor cells more efficiently than each of delivery system alone. Furthermore, these results revealed that our combined delivery platform of pTRAIL and DOX using Alkyl-modified PPI G5 can significantly improve the anti-tumor activity and this strategy might develop a new therapeutic window for cancer treatment.
Collapse
|
33
|
Beyrath J, Chekkat N, Smulski CR, Lombardo CM, Lechner MC, Seguin C, Decossas M, Spanedda MV, Frisch B, Guichard G, Fournel S. Synthetic ligands of death receptor 5 display a cell-selective agonistic effect at different oligomerization levels. Oncotarget 2018; 7:64942-64956. [PMID: 27409341 PMCID: PMC5323128 DOI: 10.18632/oncotarget.10508] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 05/11/2016] [Indexed: 01/28/2023] Open
Abstract
DR4 (Death Receptor 4) and DR5 (Death Receptor 5) are two potential targets for cancer therapy due to their ability to trigger apoptosis of cancer cells, but not normal ones, when activated by their cognate ligand TRAIL (TNF related apoptosis-inducing ligand). Therapies based on soluble recombinant TRAIL or agonist antibodies directed against one of the receptors are currently under clinical trials. However, TRAIL-R positive tumor cells are frequently resistant to TRAIL induced apoptosis. The precise mechanisms of this resistance are still not entirely understood. We have previously reported on synthetic peptides that bind to DR5 (TRAILmim/DR5) and induce tumor cell apoptosis in vitro and in vivo. Here, we showed that while hexameric soluble TRAIL is able to efficiently kill the DR5 positive lymphoma Jurkat or the carcinoma HCT116, these cells are resistant to apoptosis induced by the divalent form of TRAILmim/DR5 and are poorly sensitive to apoptosis induced by an anti-DR5 agonist monoclonal antibody. This resistance can be restored by the cross-linking of anti-DR5 agonist antibody but not by the cross-linking of the divalent form of TRAILmim/DR5. Interestingly, the divalent form of TRAILmim/DR5 that induced apoptosis of DR5 positive BJAB cells, acts as an inhibitor of TRAIL-induced apoptosis on Jurkat and HCT116 cells. The rapid internalization of DR5 observed when treated with divalent form of TRAILmim/DR5 could explain the antagonist activity of the ligand on Jurkat and HCT116 cells but also highlights the independence of the mechanisms responsible for internalization and activation when triggering the DR5 apoptotic cascade.
Collapse
Affiliation(s)
- Julien Beyrath
- Institut de Biologie Moléculaire et Cellulaire, UMR 3572, Laboratoire d'Immunopathologie et Chimie Thérapeutique, Strasbourg 67084, France.,Current address: Khondrion BV, Nijmegen 6525EX, The Netherlands
| | - Neila Chekkat
- Institut de Biologie Moléculaire et Cellulaire, UMR 3572, Laboratoire d'Immunopathologie et Chimie Thérapeutique, Strasbourg 67084, France.,Current address: Faculté de Pharmacie, UMR 7199, Laboratoire de Conception et Application de Molécules Bioactives, Illkirch BP 67401, France
| | - Cristian R Smulski
- Institut de Biologie Moléculaire et Cellulaire, UMR 3572, Laboratoire d'Immunopathologie et Chimie Thérapeutique, Strasbourg 67084, France.,Current address: University Medical Center Freiburg, Center for Chronic Immunodeficiency, Freiburg D-79110, Germany
| | - Caterina M Lombardo
- Institut Européen de Chimie et Biologie, UMR 5248, Institut de Chimie & Biologie des Membranes & des Nano-objets (CBMN), Univ. Bordeaux, Pessac 33607, France.,UMR 5248, CBMN, CNRS, Pessac 33600, France
| | - Marie-Charlotte Lechner
- Institut de Biologie Moléculaire et Cellulaire, UMR 3572, Laboratoire d'Immunopathologie et Chimie Thérapeutique, Strasbourg 67084, France.,Institut Européen de Chimie et Biologie, UMR 5248, Institut de Chimie & Biologie des Membranes & des Nano-objets (CBMN), Univ. Bordeaux, Pessac 33607, France.,UMR 5248, CBMN, CNRS, Pessac 33600, France
| | - Cendrine Seguin
- Current address: Faculté de Pharmacie, UMR 7199, Laboratoire de Conception et Application de Molécules Bioactives, Illkirch BP 67401, France
| | - Marion Decossas
- Institut de Biologie Moléculaire et Cellulaire, UMR 3572, Laboratoire d'Immunopathologie et Chimie Thérapeutique, Strasbourg 67084, France.,UMR 5248, CBMN, CNRS, Pessac 33600, France.,UMR 5248, CBMN, Univ. Bordeaux, Pessac 33600, France
| | - Maria Vittoria Spanedda
- Current address: Faculté de Pharmacie, UMR 7199, Laboratoire de Conception et Application de Molécules Bioactives, Illkirch BP 67401, France
| | - Benoît Frisch
- Current address: Faculté de Pharmacie, UMR 7199, Laboratoire de Conception et Application de Molécules Bioactives, Illkirch BP 67401, France
| | - Gilles Guichard
- Institut Européen de Chimie et Biologie, UMR 5248, Institut de Chimie & Biologie des Membranes & des Nano-objets (CBMN), Univ. Bordeaux, Pessac 33607, France.,UMR 5248, CBMN, CNRS, Pessac 33600, France
| | - Sylvie Fournel
- Institut de Biologie Moléculaire et Cellulaire, UMR 3572, Laboratoire d'Immunopathologie et Chimie Thérapeutique, Strasbourg 67084, France.,Current address: Faculté de Pharmacie, UMR 7199, Laboratoire de Conception et Application de Molécules Bioactives, Illkirch BP 67401, France
| |
Collapse
|
34
|
Deng X, Zhao W, Song L, Ying W, Guo X. Pro-apoptotic effect of TRAIL-transfected endothelial progenitor cells on glioma cells. Oncol Lett 2018; 15:5004-5012. [PMID: 29545899 PMCID: PMC5840765 DOI: 10.3892/ol.2018.7977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 01/11/2018] [Indexed: 12/14/2022] Open
Abstract
Glioma is one of the most common aggressive neuroepithelial malignant tumors in the central nervous system. It has a high recurrence rate and poor prognosis, primarily due to the fact that novel therapeutic agents cannot penetrate the blood-brain barrier (BBB). Endothelial progenitor cells (EPCs) have been reported to move across the BBB and access the tumor site. However, whether EPCs expressing the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induce glioma cell apoptosis requires further investigation. In the present study, EPCs were transfected and stably expressed with TRAIL through lentiviral infection. The pro-apoptotic effect of these TRAIL-expressing EPCs on the SHG44 glioma cell line was investigated. The migration ability of TRAIL-expressing EPCs toward SHG44 cells through the Transwell culture system was investigated via a high-content screening assay. The apoptotic rate and the expression of cleaved caspase-8 and −3 in addition to the cleaved poly(ADP-ribose) polymerase in SHG44 cells significantly increased in the TRAIL-overexpressing EPC treatment group compared with the controls. The increased apoptotic rate was reversed using a caspase inhibitor. The findings suggested that the TRAIL-expressing EPCs induced apoptosis in the SHG44 cells by activating the death receptor pathway, indicating that the TRAIL-expressing EPCs may be a useful strategy for glioma treatment.
Collapse
Affiliation(s)
- Xin Deng
- Department of Neuro-Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Wen Zhao
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China.,Co-innovation Center of Henan for New Drug R & D and Preclinical Safety; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Laijun Song
- Department of Neuro-Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Wei Ying
- Department of Neuro-Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xinbin Guo
- Department of Neuro-Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
35
|
Zhang T, Cai X, Li Q, Xue P, Chen Z, Dong X, Xue Y. Hsa-miR-875-5p exerts tumor suppressor function through down-regulation of EGFR in colorectal carcinoma (CRC). Oncotarget 2018; 7:42225-42240. [PMID: 27302926 PMCID: PMC5173130 DOI: 10.18632/oncotarget.9944] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 04/09/2016] [Indexed: 12/29/2022] Open
Abstract
Hsa-miRNA-875-5p (miR-875-5p) has recently been discovered to have anticancer efficacy in different organs. However, the role of miR-875-5p on colorectal carcinoma (CRC) is still ambiguous. In this study, we investigated the role of miR-875-5p on the development of CRC. The results indicated that miR-875-5p was significantly down-regulated in primary tumor tissues and very low levels were found in CRC cell lines. Ectopic expression of miR-875-5p in CRC cell lines significantly suppressed cell growth as evidenced by cell viability assay, colony formation assay and BrdU staining, through inhibition of cyclin D1, cyclin D2, CDK4 and up-regulation of p57(Kip2) and p21(Waf1/Cip1). In addition, miR-875-5p induced apoptosis, as indicated by concomitantly with up-regulation of key apoptosis protein cleaved caspase-3, and down-regulation of anti-apoptosis protein Bcl2. Moreover, miR-875-5p inhibited cellular migration and invasiveness through inhibition of matrix metalloproteinases (MMP)-7 and MMP-9. Further, oncogene EGFR was revealed to be a putative target of miR-875-5p, which was inversely correlated with miR-875-5p expression in CRC. Taken together, our results demonstrated that miR-875-5p played a pivotal role on CRC through inhibiting cell proliferation, migration, invasion, and promoting apoptosis by targeting oncogenic EGFR.
Collapse
Affiliation(s)
- Tiening Zhang
- Oncology Center, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200080, P. R. China
| | - Xun Cai
- Oncology Center, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200080, P. R. China
| | - Qi Li
- Oncology Center, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200080, P. R. China
| | - Peng Xue
- Oncology Center, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200080, P. R. China
| | - Zhixiao Chen
- Oncology Center, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200080, P. R. China
| | - Xiao Dong
- Oncology Center, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200080, P. R. China
| | - Ying Xue
- Oncology Center, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200080, P. R. China
| |
Collapse
|
36
|
Yang J, Liu Q, Cao S, Xu T, Li X, Zhou D, Pan L, Li C, Huang C, Meng X, Zhang L, Wang X. MicroRNA-145 Increases the Apoptosis of Activated Hepatic Stellate Cells Induced by TRAIL through NF-κB Signaling Pathway. Front Pharmacol 2018; 8:980. [PMID: 29375381 PMCID: PMC5770373 DOI: 10.3389/fphar.2017.00980] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/21/2017] [Indexed: 12/18/2022] Open
Abstract
During the liver fibrosis recovery stage tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can effectively induce apoptosis of activated hepatic stellate cells (HSCs). Normal hepatic stellate cells are resistant to TRAIL cytotoxicity. Therefore, enhancing the sensitivity of TRAIL-induced apoptosis of HSCs may be useful to treat hepatic fibrogenesis. Here, we demonstrated that miR-145 and TRAIL were down-regulated in both liver fibrosis tissue samples and transforming growth factor-β1 induced HSCs, concomitant with increased the expression of ZEB2. In addition, we found that mimics-mediated over-expression of miR-145 led to resistance to the ZEB2 expression and up-regulation of the TRAIL-induced apoptosis after treatment of LX-2 cells with TRAIL. Furthermore, ZEB2-siRNA transfected LX-2 cells showed the increased sensitivity to TRAIL-induced apoptosis. Whereas, opposite results were obtained in miR-145-inhibitor group or ZEB2 plasmid group. Moreover, miR-145 regulated ZEB2 gene expression by specifically interacting with the 3′-UTR of ZEB2 mRNA to inhibit the expression of ZEB2. Further studies showed that the over-expression of ZEB2 could inhibit TRAIL-induced apoptosis via inhibiting nuclear factor-κB (NF-κB) signaling pathway in LX-2 cells. Collectively, our data suggest that up-regulation of miR-145 can down-regulate ZEB2 expression, consequently promoting TRAIL-induced apoptosis in LX-2 cells through NF-κB signaling pathway, which facilitates the resolution of liver fibrosis.
Collapse
Affiliation(s)
- Junfa Yang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Qingxue Liu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Shiyang Cao
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Tao Xu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiaofeng Li
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Dandan Zhou
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Linxin Pan
- School of Life Sciences, Anhui Medical University, Hefei, China
| | - Changyao Li
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Cheng Huang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiaoming Meng
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Lei Zhang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiao Wang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
37
|
Yoon JY, Lee JJ, Gu S, Jung ME, Cho HS, Lim JH, Jun SY, Ahn JH, Min JS, Choi MH, Jeon SJ, Lee YJ, Go A, Heo YJ, Jung CR, Choi G, Lee K, Jeon MK, Kim NS. Novel indazole-based small compounds enhance TRAIL-induced apoptosis by inhibiting the MKK7-TIPRL interaction in hepatocellular carcinoma. Oncotarget 2017; 8:112610-112622. [PMID: 29348850 PMCID: PMC5762535 DOI: 10.18632/oncotarget.22614] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 09/29/2017] [Indexed: 01/13/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most malignant tumors. Although various treatments, such as surgery and chemotherapy, have been developed, a novel alternative therapeutic approach for HCC therapy is urgently needed. Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) is a promising anti-cancer agent, but many cancer cells are resistant to TRAIL-induced apoptosis. To help overcome TRAIL resistance in HCC cancer cells, we have identified novel chemical compounds that act as TRAIL sensitizers. We first identified the hit compound, TRT-0002, from a chemical library of 6,000 compounds using a previously developed high-throughput enzyme-linked immunosorbent assay (ELISA) screening system, which was based on the interaction of mitogen-activated protein kinase kinase 7 (MKK7) and TOR signaling pathway regulator-like (TIPRL) proteins and a cell viability assay. To increase the efficacy of this TRAIL sensitizer, we synthesized 280 analogs of TRT-0002 and finally identified two lead compounds (TRT-0029 and TRT-0173). Co-treating cultured Huh7 cells with either TRT-0029 or TRT-0173 and TRAIL resulted in TRAIL-induced apoptosis due to the inhibition of the MKK7-TIPRL interaction and subsequent phosphorylation of MKK7 and c-Jun N-terminal kinase (JNK). In vivo, injection of these compounds and TRAIL into HCC xenograft tumors resulted in tumor regression. Taken together, our results suggest that the identified lead compounds serve as TRAIL sensitizers and represent a novel strategy to overcome TRAIL resistance in HCC.
Collapse
Affiliation(s)
- Ji-Yong Yoon
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea
| | - Jeong-Ju Lee
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea
| | - Sujin Gu
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Myoung Eun Jung
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Hyun-Soo Cho
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Jung Hwa Lim
- Gene Therapy Research Unit, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Soo Young Jun
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Jun-Ho Ahn
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea
| | - Ju-Sik Min
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea
| | - Min-Hyuk Choi
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Su-Jin Jeon
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Yong-Jae Lee
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea
| | - Areum Go
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea.,Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Yun-Jeong Heo
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Cho-Rok Jung
- Gene Therapy Research Unit, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Gildon Choi
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea.,Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Kwangho Lee
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea.,Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Moon-Kook Jeon
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Nam-Soon Kim
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| |
Collapse
|
38
|
Enhancement of TRAIL-induced apoptosis by 5-fluorouracil requires activating Bax and p53 pathways in TRAIL-resistant lung cancers. Oncotarget 2017; 8:18095-18105. [PMID: 28178647 PMCID: PMC5392310 DOI: 10.18632/oncotarget.14994] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 01/03/2017] [Indexed: 12/14/2022] Open
Abstract
Lung cancer, especially lung adenocarcinoma, is one of the main causes of death worldwide. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a primary anticancer agent and a member of the tumor necrosis factor family that selectively induces apoptosis in various tumor cells, but not in normal cells. Combination chemotherapy can be used for treating specific cancer types even at progressive stages. In the present study, we observed that 5-fluorouracil, which exerts anticancer effects by inhibiting tumor cell proliferation, enhanced TRAIL-induced apoptosis of TRAIL-resistant human adenocarcinoma A549 cells. Interestingly, 5-fluorouracil treatment markedly increased Bax and p53 levels and 5-fluorouracil and TRAIL cotreatment increased Ac-cas3 and Ac-cas8 levels compared with those in control cells. Taken together, the present study demonstrated that 5-fluorouracil enhances TRAIL-induced apoptosis in TRAIL-resistant lung adenocarcinoma cells by activating Bax and p53, and also suggest that TRAIL and 5-fluorouracil cotreatment can be used as an adequate therapeutic strategy for TRAIL-resistant human cancers.
Collapse
|
39
|
Yang SZ, Xu F, Zhou T, Zhao X, McDonald JM, Chen Y. The long non-coding RNA HOTAIR enhances pancreatic cancer resistance to TNF-related apoptosis-inducing ligand. J Biol Chem 2017; 292:10390-10397. [PMID: 28476883 PMCID: PMC5481552 DOI: 10.1074/jbc.m117.786830] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/21/2017] [Indexed: 08/11/2023] Open
Abstract
Pancreatic cancer is a malignant neoplasm with a high mortality rate. Therapeutic agents that activate TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis have shown promising efficacy, but many pancreatic cancers are resistant to TRAIL therapy. Epigenetic regulation plays important roles in tumor pathogenesis and resistance, and a recent study indicated that the long non-coding RNA HOX transcript antisense RNA (HOTAIR) is overexpressed in pancreatic cancer. However, the role of HOTAIR in pancreatic cancer resistance to anticancer agents is unknown. The present study determined the role of HOTAIR in pancreatic cancer TRAIL resistance and investigated the underlying molecular mechanisms. We observed that TRAIL-resistant pancreatic cancer cells had higher levels of HOTAIR expression, whereas TRAIL-sensitive pancreatic cancer cells had lower HOTAIR levels. Overexpressing HOTAIR in TRAIL-sensitive cells attenuated TRAIL-induced apoptosis, and shRNA-mediated HOTAIR knockdown in TRAIL-resistant PANC-1 cells sensitized them to TRAIL-induced apoptosis. These results support a causative effect of HOTAIR on TRAIL sensitivity. Mechanistically, we found that increased HOTAIR expression inhibited the expression of the TRAIL receptor death receptor 5 (DR5), whereas HOTAIR knockdown increased DR5 expression. We further demonstrated that HOTAIR regulates DR5 expression via the epigenetic regulator enhancer of zeste homolog 2 (EZH2) and that EZH2 controls histone H3 lysine 27 trimethylation on the DR5 gene. Taken together, these results demonstrate that high HOTAIR levels increase the resistance of pancreatic cancer cells to TRAIL-induced apoptosis via epigenetic regulation of DR5 expression. Our study therefore supports the notion that targeting HOTAIR function may represent a strategy to overcome TRAIL resistance in pancreatic cancer.
Collapse
Affiliation(s)
| | - Fei Xu
- From the Departments of Pathology
| | | | - Xinyang Zhao
- Biochemistry, University of Alabama at Birmingham and
| | - Jay M McDonald
- From the Departments of Pathology
- the Research Department, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama 35294
| | - Yabing Chen
- From the Departments of Pathology,
- the Research Department, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama 35294
| |
Collapse
|
40
|
Dai W, Wang X, Song G, Liu T, He B, Zhang H, Wang X, Zhang Q. Combination antitumor therapy with targeted dual-nanomedicines. Adv Drug Deliv Rev 2017; 115:23-45. [PMID: 28285944 DOI: 10.1016/j.addr.2017.03.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/24/2017] [Accepted: 03/03/2017] [Indexed: 01/01/2023]
Abstract
Combination therapy is one of the important treatment strategies for cancer at present. However, the outcome of current combination therapy based on the co-administration of conventional dosage forms is suboptimal, due to the short half-lives of chemodrugs, their deficient tumor selectivity and so forth. Nanotechnology-based targeted delivery systems show great promise in addressing the associated problems and providing superior therapeutic benefits. In this review, we focus on the combination of therapeutic strategies between different nanomedicines or drug-loaded nanocarriers, rather than the co-delivery of different drugs via a single nanocarrier. We introduce the general concept of various targeting strategies of nanomedicines, present the principles of combination antitumor therapy with dual-nanomedicines, analyze their advantages and limitations compared with co-delivery strategies, and overview the recent advances of combination therapy based on targeted nanomedicines. Finally, we reviewed the challenges and future perspectives regarding the selection of therapeutic agents, targeting efficiency and the gap between the preclinical and clinical outcome.
Collapse
Affiliation(s)
- Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiaoyou Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Ge Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Tongzhou Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China.
| |
Collapse
|
41
|
Beyer K, Partecke LI, Roetz F, Fluhr H, Weiss FU, Heidecke CD, von Bernstorff W. LPS promotes resistance to TRAIL-induced apoptosis in pancreatic cancer. Infect Agent Cancer 2017; 12:30. [PMID: 28572836 PMCID: PMC5450120 DOI: 10.1186/s13027-017-0139-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 05/16/2017] [Indexed: 12/29/2022] Open
Abstract
Background Though TRAIL has been hailed as a promising drug for tumour treatment, it has been observed that many tumour cells have developed escape mechanisms against TRAIL-induced apoptosis. As a receptor of LPS, TLR 4, which is expressed on a variety of cancer cells, can be associated with TRAIL-resistance of tumour cells and tumour progression as well as with the generation of an anti-tumour immune response. Methods In this study, the sensitivity to TRAIL-induced apoptosis as well as the influence of LPS-co-stimulation on the cell viability of the pancreatic cancer cell lines PANC-1, BxPC-3 and COLO 357 was examined by FACS analyses and a cell viability assay. Subsequently, the expression of TRAIL-receptors was detected via FACS analyses. Levels of osteoprotegerin (OPG) were also determined using an enzyme-linked immunosorbent assay. Results PANC-1 cells were shown to be resistant to TRAIL-induced apoptosis. This was accompanied by significantly increased osteoprotegerin levels and a significantly decreased expression of DR4. In contrast, TRAIL significantly induced apoptosis in COLO 357 cells and to a lesser degree in BxPC-3 cells. Co-stimulation of COLO 357 as well as BxPC-3 cells combining TRAIL and LPS resulted in a significant decrease in TRAIL-induced apoptosis. In COLO 357 cells TRAIL-stimulation decreased the levels of OPG thereby not altering the expression of the TRAIL-receptors 1–4 resulting in a high susceptibility to TRAIL-induced apoptosis. Co-stimulation with LPS and TRAIL completely reversed the effect of TRAIL on OPG levels reaching a 2-fold increase beyond the level of non-stimulated cells resulting in a lower susceptibility to apoptosis. In BxPC-3, TRAIL stimulation decreased the expression of DR4 and significantly increased the decoy receptors TRAIL-R3 and TRAIL-R4 leading to a decrease in TRAIL-induced apoptosis. OPG levels remained unchanged. Co-stimulation with TRAIL and LPS further enhanced the changes in TRAIL-receptor-expression promoting apoptosis resistance. Conclusions Here it has been shown that TRAIL-resistance in pancreatic cancer cells can be mediated by the inflammatory molecule LPS as well as by different expression patterns of functional and non-functional TRAIL-receptors.
Collapse
Affiliation(s)
- Katharina Beyer
- Department of General, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, Greifswald, Germany.,Department of General, Visceral and Vascular Surgery, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Lars Ivo Partecke
- Department of General, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Felicitas Roetz
- Department of General, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Herbert Fluhr
- Department of Medicine A, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Frank Ulrich Weiss
- Department of Obstetrics and Gynaecology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Claus-Dieter Heidecke
- Department of General, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Wolfram von Bernstorff
- Department of General, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
42
|
Mielczarek-Palacz A, Sikora J, Kondera-Anasz Z. Assessment of concentrations of sTRAIL ligand and its receptors sTRAIL-R1 and sTRAIL-R2 - markers monitoring the course of the extrinsic pathway of apoptosis induction: potential application in ovarian cancer diagnostics. Arch Med Sci 2017; 13:624-628. [PMID: 28507579 PMCID: PMC5420616 DOI: 10.5114/aoms.2015.53144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 06/22/2015] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION TNF-related apoptosis-inducing ligand (TRAIL) together with its receptors are involved in activation of the extrinsic pathway of apoptosis. Due to the special role of the apoptosis pathway in pathogenesis of ovarian cancers, the aim of the study was to assess concentrations of sTRAIL, sTRAIL-R1 and sTRAIL-R2 in serum of affected women. MATERIAL AND METHODS The study group included 85 women with diagnosed ovarian tumors: 35 women with ovarian serous cystadenoma, 15 women with ovarian teratoma and 35 women with serous cystadenocarcinoma. The control group consisted of 30 healthy women. Concentrations of studied parameters were measured by ELISA methods. RESULTS Serum levels of all studied parameters were higher in serum of women with ovarian tumors than in the controls, but their concentrations varied depending on the clinical diagnosis. The highest concentration of TRAIL was found in serum of women with ovarian cancer, the highest sTRAIL-R1 level in serum of women with ovarian mature teratoma, and the highest sTRAIL-R2 level in serum of women with ovarian serous cystadenoma. CONCLUSIONS The state of immunosuppression accompanying neoplastic disease depends on the extrinsic pathway of apoptosis induction in the TRAIL/TRAIL-R system. Determination of TRAIL-R1 and TRAIL-R2 levels may prove to be useful in ovarian tumor differential diagnostics, which requires further research.
Collapse
Affiliation(s)
- Aleksandra Mielczarek-Palacz
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Immunology and Serology, Medical University of Silesia, Katowice, Sosnowiec, Poland
| | - Justyna Sikora
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Immunology and Serology, Medical University of Silesia, Katowice, Sosnowiec, Poland
| | - Zdzisława Kondera-Anasz
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Immunology and Serology, Medical University of Silesia, Katowice, Sosnowiec, Poland
| |
Collapse
|
43
|
Wu Y, He J, Geng J, An Y, Ye X, Yan S, Yu Q, Yin J, Zhang Z, Li D. Recombinant Newcastle disease virus expressing human TRAIL as a potential candidate for hepatoma therapy. Eur J Pharmacol 2017; 802:85-92. [PMID: 28246027 DOI: 10.1016/j.ejphar.2017.02.042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 02/22/2017] [Accepted: 02/24/2017] [Indexed: 01/21/2023]
Abstract
Newcastle disease virus (NDV) have shown oncolytic therapeutic efficacy in preclinical studies and are currently proved for clinical trials. We have previously reported, for the first time, NDV Anhinga strain has an efficient cancer therapeutic efficacy in hepatoma. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) functions as a cytokine to selectively kill various cancer cells without toxicity to most normal cells. Numerous studies have demonstrated the potential use of recombinant soluble TRAIL as a cancer therapeutic agent. In this study, we have showed administration of a recombinant NDV Anhinga strain expressing soluble TRAIL (NDV/Anh-TRAIL) results in an efficient suppression of hepatocellular carcinoma without significant toxicity. The results show that recombinant NDV Anhinga strain expressing soluble TRAIL is a promising candidate for hepatoma therapy.
Collapse
Affiliation(s)
- Yunzhou Wu
- College of Life Science, Northeast Agriculture University, Mucai Street 59, Xiangfang District, Harbin, China
| | - Jinjiao He
- College of life science and technology, Xinxiang University, Jinsui Avenue, Hongqi District, Xinxiang, China
| | - Jingshu Geng
- The Pathology Department, Affiliated Tumor Hospital of Harbin Medical University, Yiyuan Street 37, Nangang District, Harbin, China
| | - Ying An
- College of Life Science, Northeast Agriculture University, Mucai Street 59, Xiangfang District, Harbin, China
| | - Xianlong Ye
- School of Life Science, Henan Normal University, 46 Jianshe Road E., Xinxiang, China
| | - Shijun Yan
- College of Life Science, Northeast Agriculture University, Mucai Street 59, Xiangfang District, Harbin, China
| | - Qingzhong Yu
- Southeast Poultry Research Laboratory, Agricultural Research Service, United States Department of Agriculture, 934 College Station Road, Athens, GA 30605, USA
| | - Jiechao Yin
- College of Life Science, Northeast Agriculture University, Mucai Street 59, Xiangfang District, Harbin, China
| | - Zhenyu Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Deshan Li
- College of Life Science, Northeast Agriculture University, Mucai Street 59, Xiangfang District, Harbin, China
| |
Collapse
|
44
|
Li J, Wang Y, Liu L, Yuan Y, Bao Y. [Thioridazine Sensitizes Apoptotic Effect of TRAIL in Human Lung Cancer PC9 Cells Through ER Stress Mediated Up-regulation of DR5]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2017; 20:80-87. [PMID: 28228218 PMCID: PMC5972967 DOI: 10.3779/j.issn.1009-3419.2017.02.02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
背景与目的 肿瘤坏死因子相关凋亡诱导配体(tumor necrosis factor-related apoptosis-inducting ligand, TRAIL)可诱导肿瘤细胞发生凋亡,然而相当数量的肿瘤细胞可耐受TRAIL诱导的凋亡而得以存活。本实验观察硫利达嗪(thioridazine, THZ)通过诱导内质网应激(endoplasmic reticulum stress, ER stress)介导的死亡受体5(death receptor 5, DR5)表达上调,继而增敏TRAIL对肺腺癌细胞PC9的生长抑制及凋亡诱导效应,探讨其机制。 方法 不同浓度硫利达嗪及TRAIL单独或联合处理PC9细胞,MTT法检测细胞活性变化,流式细胞术检测细胞表面DR5表达及细胞凋亡率,Western blotting检测内质网应激相关蛋白GRP78(glucose regulated protein 78)、C/EBP环磷酸腺苷反应元件结合转录因子同源蛋白(C/EBP homologous protein, CHOP)、p-PERK(PKR-like ER kinase)、p-eIF2α(eukaryotic initiation factor-2α, eIF2α)、ATF4(activating transcription factor 4, ATF4)及凋亡相关蛋白Caspase-3、Caspase-9、Caspase-8、PARP、DR5表达变化。 结果 硫利达嗪对PC9细胞的增殖抑制效应呈浓度依赖性(P < 0.05),硫利达嗪可增加TRAIL对PC9细胞的抑制作用及凋亡诱导作用且可上调PC9细胞表面DR5表达水平,流式细胞术结果显示:TRAIL联合硫利达嗪组细胞凋亡率较单独TRAIL组显著增加(P < 0.05),Western blotting结果显示:TRAIL联合硫利达嗪组细胞Cleaved-caspase-8、Cleaved-PARP、DR5表达水平较单独TRAIL组明显上调。DR5表达上调及促凋亡效应是通过诱导内质网应激发生,并伴随着GRP78及CHOP表达上调发生的,且该效应可被4-苯基丁酸(4-phenylbutyric acid, 4-PBA)可抑制(P < 0.05)。 结论 硫利达嗪增敏TRAIL对PC9细胞的增殖抑制效应显著,其机制可能与硫利达嗪内质网应激介导的DR5上调有关。
Collapse
Affiliation(s)
- Juan Li
- Department of Oncology, the Third Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Yi Wang
- Department of Oncology, the Third Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Liu Liu
- Department of Oncology, the Third Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Yuan Yuan
- Central Laboratory of Hefei Binhu Hospital, Hefei 230061, China
| | - Yangyi Bao
- Department of Oncology, the Third Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| |
Collapse
|
45
|
Sun C, Huang C, Li S, Yang C, Xi Y, Wang L, Zhang F, Fu Y, Li D. Hsa-miR-326 targets CCND1 and inhibits non-small cell lung cancer development. Oncotarget 2016; 7:8341-59. [PMID: 26840018 PMCID: PMC4884997 DOI: 10.18632/oncotarget.7071] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 01/13/2016] [Indexed: 12/12/2022] Open
Abstract
Hsa-miRNA-326 (miR-326) has recently been discovered having anticancer efficacy in different organs. However, the role of miR-326 on non-small cell lung cancer (NSCLC) is still ambiguous. In this study, we investigated the role of miR-326 on the development of NSCLC. The results indicated that miR-326 was significantly down-regulated in primary tumor tissues and very low levels were found in NSCLC cell lines. Ectopic expression of miR-326 in NSCLC cell lines significantly suppressed cell growth as evidenced by cell viability assay, colony formation assay and BrdU staining, through inhibition of cyclin D1, cyclin D2, CDK4 and up-regulation of p57(Kip2) and p21(Waf1/Cip1). In addition, miR-326 induced apoptosis, as indicated by concomitantly with up-regulation of key apoptosis protein cleaved caspase-3, and down-regulation of anti-apoptosis protein Bcl2. Moreover, miR-326 inhibited cellular migration and invasiveness through inhibition of matrix metalloproteinases (MMP)-7 and MMP-9. Further, oncogene CCND1 was revealed to be a putative target of miR-326, which was inversely correlated with miR-326 expression in NSCLC. Taken together, our results demonstrated that miR-326 played a pivotal role on NSCLC through inhibiting cell proliferation, migration, invasion, and promoting apoptosis by targeting oncogenic CCND1.
Collapse
Affiliation(s)
- Chengcao Sun
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
| | - Chuanfeng Huang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China.,Department of Pharmacology, Basic Medical School, Nanyang Medical College, 473003 Nanyang, P.R.China
| | - Shujun Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China.,Wuhan Hospital for The Prevention and Treatment of Occupational Diseases, 430071 Wuhan, P.R.China
| | - Cuili Yang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
| | - Yongyong Xi
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
| | - Liang Wang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
| | - Feng Zhang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
| | - Yunfeng Fu
- The Third Xiang-Ya Hospital, Central South University, 410013 Changsha, P.R.China
| | - Dejia Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
| |
Collapse
|
46
|
Zhu J, Zhou Q, Tan S. Targeting miRNAs associated with surface expression of death receptors to modulate TRAIL resistance in breast cancer. Cancer Lett 2016; 383:154-160. [PMID: 27693456 DOI: 10.1016/j.canlet.2016.09.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 08/22/2016] [Accepted: 09/02/2016] [Indexed: 01/07/2023]
Abstract
Tumor necrosis factor related apoptosis-inducing ligand (TRAIL) is capable of inducing apoptosis upon engagement of its death receptors (DRs) 4 and 5. TRAIL therapy has garnered intense interest as one of the most promising agents for cancer therapy, for its selective induction of tumor-cell apoptosis while low toxicity to most normal cells. However, a variety of breast cancer cell lines could be resistant to TRAIL-induced apoptosis. Absence of DR4 and DR5 on the breast cancer cell surface has been proposed to be critically involved in resistance to TRAIL and its agonistic antibodies. Moreover, endocytosis and autophagy in breast cancer cells could induce TRAIL resistance through downregulation of surface DR4/5. MicroRNAs (miRNAs), as endogenously expressed small non-coding RNAs, function as regulators of gene expression and involve tremendous biological processes including drug resistance. In this review, we highlight recent advances in the functional role of miRNAs in endocytosis and autophagy pathways. This review aims to present that, through regulation of critical molecules involved in autophagy and endocytosis, miRNAs could lead to mislocalization of DR4/5 in breast cancer cells and therefore play an important role in TRAIL-mediated apoptosis and TRAIL resistance.
Collapse
Affiliation(s)
- Juanjuan Zhu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Qiujing Zhou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Shuhua Tan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
47
|
Voltan R, Secchiero P, Casciano F, Milani D, Zauli G, Tisato V. Redox signaling and oxidative stress: Cross talk with TNF-related apoptosis inducing ligand activity. Int J Biochem Cell Biol 2016; 81:364-374. [PMID: 27686849 DOI: 10.1016/j.biocel.2016.09.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/21/2016] [Accepted: 09/24/2016] [Indexed: 02/06/2023]
Abstract
Redox regulation plays a key role in several physiopathological contexts and free radicals, from nitric oxide and superoxide anion up to other forms of reactive oxygen species (ROS), have been demonstrated to be involved in different biological and regulatory processes. The data reported in the current literature describe a link between ROS, inflammation and programmed cell death that is attracting interest as new pathways to be explored and targeted for therapeutic purposes. In this light, there is also growing attention to the involvement of this link in the activity of the TNF-related apoptosis inducing ligand (TRAIL). TRAIL is a member of the TNF ligands super family able to mediate multiple intracellular signals, with the potential to lead to a range of biological effects in different cell types. In particular, the hallmark of TRAIL is the ability to induce selective apoptosis in transformed cells leaving normal cells almost unaffected and this feature has already opened the door to several clinical studies for cancer treatment. Moreover, TRAIL plays a role in several physiological and pathological processes of both innate and adaptive immune systems and of the cardiovascular context, with a strong clinical potential. Nonetheless, several issues still need to be clarified about the signaling mediated by TRAIL to gain deeper insight into its therapeutic potential. In this light, the aim of this review is to summarize the main preclinical evidences about the interplay between TRAIL and redox signaling, with particular emphasis to the implications in vascular physiopathology and cancer.
Collapse
Affiliation(s)
- Rebecca Voltan
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy
| | - Paola Secchiero
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy
| | - Fabio Casciano
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy
| | - Daniela Milani
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy
| | - Veronica Tisato
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy.
| |
Collapse
|
48
|
Redza-Dutordoir M, Averill-Bates DA. Activation of apoptosis signalling pathways by reactive oxygen species. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2977-2992. [PMID: 27646922 DOI: 10.1016/j.bbamcr.2016.09.012] [Citation(s) in RCA: 2380] [Impact Index Per Article: 264.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 09/12/2016] [Accepted: 09/15/2016] [Indexed: 02/08/2023]
Abstract
Reactive oxygen species (ROS) are short-lived and highly reactive molecules. The generation of ROS in cells exists in equilibrium with a variety of antioxidant defences. At low to modest doses, ROS are considered to be essential for regulation of normal physiological functions involved in development such as cell cycle progression and proliferation, differentiation, migration and cell death. ROS also play an important role in the immune system, maintenance of the redox balance and have been implicated in activation of various cellular signalling pathways. Excess cellular levels of ROS cause damage to proteins, nucleic acids, lipids, membranes and organelles, which can lead to activation of cell death processes such as apoptosis. Apoptosis is a highly regulated process that is essential for the development and survival of multicellular organisms. These organisms often need to discard cells that are superfluous or potentially harmful, having accumulated mutations or become infected by pathogens. Apoptosis features a characteristic set of morphological and biochemical features whereby cells undergo a cascade of self-destruction. Thus, proper regulation of apoptosis is essential for maintaining normal cellular homeostasis. ROS play a central role in cell signalling as well as in regulation of the main pathways of apoptosis mediated by mitochondria, death receptors and the endoplasmic reticulum (ER). This review focuses on current understanding of the role of ROS in each of these three main pathways of apoptosis. The role of ROS in the complex interplay and crosstalk between these different signalling pathways remains to be further unravelled during the coming years.
Collapse
Affiliation(s)
- Maureen Redza-Dutordoir
- Département des Sciences Biologiques (TOXEN, BIOMED), Université du Québec à Montréal, Montréal, Québec, Canada
| | - Diana A Averill-Bates
- Département des Sciences Biologiques (TOXEN, BIOMED), Université du Québec à Montréal, Montréal, Québec, Canada.
| |
Collapse
|
49
|
Kim H, Shin EA, Kim CG, Lee DY, Kim B, Baek NI, Kim SH. Obovatol Induces Apoptosis in Non-small Cell Lung Cancer Cells via C/EBP Homologous Protein Activation. Phytother Res 2016; 30:1841-1847. [PMID: 27489231 DOI: 10.1002/ptr.5690] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/03/2016] [Accepted: 07/11/2016] [Indexed: 12/27/2022]
Abstract
Although obovatol, a phenolic compound from the bark of Magnolia obovata, was known to have antioxidant, neuroprotective, antiinflammatory, antithrombotic and antitumour effects, its underlying antitumour mechanism is poorly understood so far. Thus, in the present study, the antitumour molecular mechanism of obovatol was investigated in non-small cell lung cancer cells (NSCLCs). Obovatol exerted cytotoxicity in A549 and H460 NSCLCs, but not in BEAS-2B cells. Also, obovatol increased sub-G1 accumulation and early and late apoptotic portion in A549 and H460 NSCLCs. Consistently, obovatol cleaved PARP, activated caspase 9/3 and Bax and attenuated the expression of cyclin D1 in A549 and H460 NSCLCs. Interestingly, obovatol upregulated the expression of endoplasmic reticulum stress proteins such as C/EBP homologous protein (CHOP), IRE1α, ATF4 and p-elF2 in A549 and H460 NSCLCs. Conversely, depletion of CHOP blocked the apoptotic activity of obovatol to increase sub-G1 accumulation in A549 and H460 NSCLCs. Overall, our findings support scientific evidences that obovatol induces apoptosis via CHOP activation in A549 and H460 NSCLCs. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Heejeong Kim
- Department of East West Medical Science, Graduate School of East West Medical Science, Kyung Hee University, Yongin, 446-701, Korea
| | - Eun Ah Shin
- College of Korean Medicine, Kyung Hee University, Seoul, 131-701, Korea
| | - Chang Geun Kim
- College of Korean Medicine, Kyung Hee University, Seoul, 131-701, Korea
| | - Dae Young Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration (RDA), Eumseong, 27709, Korea
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Seoul, 131-701, Korea
| | - Nam-In Baek
- Department of Oriental Medicine Biotechnology, Graduate School of Biotechnology, Kyung Hee University, Yongin, 446-701, Korea
| | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Seoul, 131-701, Korea.
| |
Collapse
|
50
|
Sun JG, Ruan F, Zeng XL, Xiang J, Li X, Wu P, Fung KP, Liu FY. Clitocine potentiates TRAIL-mediated apoptosis in human colon cancer cells by promoting Mcl-1 degradation. Apoptosis 2016; 21:1144-57. [DOI: 10.1007/s10495-016-1273-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|