1
|
Haghir-Sharif-Zamini Y, Khosravi A, Hassan M, Zarrabi A, Vosough M. c-FLIP/Ku70 complex; A potential molecular target for apoptosis induction in hepatocellular carcinoma. Arch Biochem Biophys 2025; 765:110306. [PMID: 39818348 DOI: 10.1016/j.abb.2025.110306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignancies worldwide and the most common form of liver cancer. Despite global efforts toward early diagnosis and effective treatments, HCC is often diagnosed at advanced stages, where conventional therapies frequently lead to resistance and/or high recurrence rates. Therefore, novel biomarkers and promising medications are urgently required. Epi-drugs, or epigenetic-based medicines, have recently emerged as a promising therapeutic modality. Since the epigenome of the cancer cells is always dysregulated and this is followed by apoptosis-resistance, reprogramming the epigenome of cancer cells by epi-drugs (such as HDAC inhibitors (HDACis), and DNMT inhibitors (DNMTis)) could be an alternative approach to use in concert with established treatment protocols. C-FLIP, an anti-apoptotic protein, and Ku70, a member of the DNA repair system, bind together and make a cytoplasmic complex in certain cancers and induce resistance to apoptosis. Many epi-drugs, such as HDACis, can dissociate this complex through Ku70 acetylation and activate cellular apoptosis. The novel compounds for dissociating this complex could provide an innovative insight into molecular targeted HCC treatments. In this review, we address the innovative therapeutic potential of targeting c-FLIP/Ku70 complex by epi-drugs, particularly HDACis, to overcome apoptosis resistance of HCC cells. This review will cover the mechanisms by which the c-FLIP/Ku70 complex facilitates cancer cell survival, the impact of epigenetic alterations on the complex dissociation, and highlight HDACis potential in combination therapies, biomarker developments and mechanistic overviews. This review highlights c-FLIP ubiquitination and Ku70 acetylation levels as diagnostic and prognostic tools in HCC management.
Collapse
Affiliation(s)
- Yasamin Haghir-Sharif-Zamini
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul, 34959, Turkiye
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, 320315, Taiwan; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai - 600 077, India.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
2
|
Li GJ, Wang C, Wang WD, Shang Y, Zeng CY, Wang AM, Bai JL, Su J, Su L, Si SY, Yu LY, Gan ML, Chen SZ. Chromomycins from soil-derived Streptomyces sp. inhibit the growth of human non-small cell lung cancer cells by targeting c-FLIP. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025; 27:153-168. [PMID: 38975979 DOI: 10.1080/10286020.2024.2375288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/09/2024]
Abstract
Three chromomycin derivatives, chromomycins A3 (1, CA3), A5 (2, CA5), and monodeacetylchromomycin A3 (3, MDA-CA3), were identified from the soil-derived Streptomyces sp. CGMCC 26516. A reinvestigation of the structure of CA5 is reported, of which the absolute configuration was unambiguously determined for the first time to be identical with that of CA3 based on nuclear magnetic resonance (NMR) data analysis as well as NMR and electronic circular dichroism calculations. Compounds 1-3 showed potent cytotoxicity against the non-small-cell lung cancer (NSCLC) cells (A549, H460, H157-c-FLIP, and H157-LacZ) and down-regulated the protein expression of c-FLIP in A549 cells. The IC50 values of chromomycins in H157-c-FLIP were higher than that in H157-LacZ. Furthermore, si-c-FLIP promoted anti-proliferation effect of chromomycins in NSCLC cells. In nude mice xenograft model, 1 and 2 both showed more potent inhibition on the growth of H157-lacZ xenografts than that of H157-c-FLIP xenografts. These results verify that c-FLIP mediates the anticancer effects of chromomycins in NSCLC.
Collapse
Affiliation(s)
- Gao-Jie Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Chen Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Wen-Die Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yue Shang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Chao-Yang Zeng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ai-Min Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jing-Lin Bai
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jing Su
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ling Su
- School of Life Sciences, Shandong University School of Life Sciences, Jinan 250100, China
| | - Shu-Yi Si
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Li-Yan Yu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Mao-Luo Gan
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shu-Zhen Chen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesInstitute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, , Beijing 100050, China
| |
Collapse
|
3
|
Guo R, Wei Y, Du Y, Liu L, Zhang H, Ren R, Sun R, Zhang T, Xiong X, Zhao L, Wang H, Guo X, Zhu X. EX527, a sirtuins 1 inhibitor, sensitizes T-cell leukemia to death receptor-mediated apoptosis by downregulating cellular FLICE inhibitory protein. Cancer Biol Ther 2024; 25:2402588. [PMID: 39286953 PMCID: PMC11409494 DOI: 10.1080/15384047.2024.2402588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/22/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Death receptor-mediated extrinsic apoptosis system had been developed as a promising therapeutic strategy in clinical oncology, such as TRAIL therapy. However, multiple studies have demonstrated that TRAIL resistance is the biggest problem for disappointing clinical trials despite preclinical success. Targeting cellular FLICE inhibitory protein (cFLIP) is one strategy of combinatorial therapies to overcome resistance to DR-mediated apoptosis due to its negative regulator of extrinsic apoptosis. E × 527 (Selisistat) is a specific inhibitor of SIRT1 activity with safe and well tolerance in clinical trials. Here, we show that E × 527 could strengthen significantly activation of rhFasL-mediated apoptotic signaling pathway and increased apoptotic rate of T leukemia cells with high expression of cFLIP. Mechanically, Inhibition of SIRT1 by E × 527 increased polyubiquitination level of cFLIP via increasing acetylation of Ku70, which could promote proteosomal degradation of cFLIP protein. It implied that combinatorial therapies of E × 527 plus TRAIL may have a potential as a novel clinical application for TRAIL-resistant hematologic malignancies.
Collapse
Affiliation(s)
- Rongqi Guo
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Yihui Wei
- Henan Red Cross Blood Center, Xinxiang, China
| | - Yating Du
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Luyue Liu
- Departments of Laboratory Medicine, Zhoukou Central Hospital, Zhoukou, China
| | - Haoqi Zhang
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang
Medical University, Xinxiang, China
| | - Ruiying Ren
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Ruili Sun
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Tingting Zhang
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang
Medical University, Xinxiang, China
| | - Xiwen Xiong
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Lijun Zhao
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Hongfei Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Xiaofang Guo
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang
Medical University, Xinxiang, China
| | - Xiaofei Zhu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
4
|
Ebrahimi K, Bagheri R, Gholamhosseinian H, Keramati MR, Rafatpanah H, Iranshahi M, Rassouli FB. Umbelliprenin improved anti-proliferative effects of ionizing radiation on adult T-cell leukemia/lymphoma cells via interaction with CDK6; an in vitro and in silico study. Int J Immunopathol Pharmacol 2024; 38:3946320241287873. [PMID: 39313767 PMCID: PMC11437583 DOI: 10.1177/03946320241287873] [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: 09/25/2024] Open
Abstract
Adult T-cell leukemia/lymphoma (ATL) is an aggressive malignancy with poor survival rates. The efficacy of radiotherapy in ATL needs enhancement with radiosensitizing agents. This study investigated whether umbelliprenin (UMB) could improve the therapeutic effects of ionizing radiation (IR) in ATL cells. UMB, a naturally occurring prenylated coumarin, exhibits anticancer properties and has shown synergistic effects when combined with chemotherapeutic drugs. Despite this promising profile, there is a notable lack of research on its potential combinatorial effects with IR, particularly for ATL treatment. UMB was extracted from Ferula persica using thin layer chromatography. MT-2 cells were treated with UMB alone and in combination with various doses of IR, and cell proliferation was assessed via alamarBlue assay. Flow cytometry with annexin V and PI staining was conducted, and candidate gene expression was analyzed by qPCR. In silico analysis involved identifying pathogenic targets of ATL, constructing protein-protein interaction (PPI) networks, and evaluating CDK6 expression in MT-2 cells. Molecular docking was used to determine the interaction between UMB and CDK6. The alamarBlue assay and flow cytometry showed that pretreating ATL cells with UMB significantly (p < .0001) enhanced anti-proliferative effects of IR. The combination index indicated a synergistic effect between UMB and IR. qPCR revealed significant (p < .0001) downregulation of CD44, CDK6, c-MYC, and cFLIPL, and overexpression of cFLIPS. Computational analysis identified CDK6 as a hub gene in the PPI network, and CDK6 overexpression was confirmed in MT-2 cells. Molecular docking revealed a favorable binding interaction between UMB and the ATP-binding site of CDK6, with a JAMDA score of -2.131, surpassing the control selonsertib. The current study provides evidence that UMB enhances the anti-proliferative effects of IR on ATL cells, and highlights the significance of targeting CDK6 in combinatorial approaches.
Collapse
Affiliation(s)
- Keyhan Ebrahimi
- Cancer Molecular Pathology Research Center, Department of Hematology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Bagheri
- Cancer Molecular Pathology Research Center, Department of Hematology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Gholamhosseinian
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Keramati
- Cancer Molecular Pathology Research Center, Department of Hematology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Immunology Research Center, Inflammation and inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh B Rassouli
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
5
|
Rafatpanah H, Golizadeh M, Mahdifar M, Mahdavi S, Iranshahi M, Rassouli FB. Conferone, a coumarin from Ferula flabelliloba, induced toxic effects on adult T-cell leukemia/lymphoma cells. Int J Immunopathol Pharmacol 2023; 37:3946320231197592. [PMID: 37688389 PMCID: PMC10493046 DOI: 10.1177/03946320231197592] [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] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Adult T-cell leukemia/lymphoma (ATL) is a lymphoid malignancy caused by HTLV-1 infection, with distinct geographical distribution. Despite advances in cancer treatment, the average survival rate of ATL is low. Conferone is a natural coumarin extracted from Ferula species with a wide range of pharmaceutical effects. In search for a novel chemotherapeutic agent, we investigated the cytotoxicity of conferone on ATL cells. METHODS To obtain conferone, the methanolic extract of the roots of F. flabelliloba was subjected to silica gel column chromatography, followed by 1H- and 13C-NMR to confirm its structure. For cytotoxicity assay, MT-2 cells were treated with different concentrations of conferone (2.5, 5, 10, 20, and 40 µM) for 24, 48, and 72 h, and viability was evaluated by a colorimetric assay using alamarBlue. Cell cycle was analyzed by PI staining and flow cytometry, and qPCR was used to study the expression of candidate genes. RESULTS AND CONCLUSION Obtained findings indicated that conferone induced considerable cytotoxic effects on MT-2 cells in a time- and dose-dependent manner. In addition, accumulation of cells in the sub-G1 phase of the cell cycle was detected upon conferone administration. Moreover, conferone reduced the expression of CDK6, c-MYC, CFLIPL, and NF-κB (Rel-A) in MT-2 cells. Accordingly, conferone could be considered as a potent agent against ATL, although complementary investigations are required to define more precisely its mechanism of action.
Collapse
Affiliation(s)
- Houshang Rafatpanah
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marziyeh Golizadeh
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Mahdifar
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shakiba Mahdavi
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh B Rassouli
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
6
|
Fornalski KW, Adamowski Ł, Bugała E, Jarmakiewicz R, Kirejczyk M, Kopyciński J, Krasowska J, Kukulski P, Piotrowski Ł, Ponikowska J, Reszczyńska J, Słonecka I, Wysocki P, Dobrzyński L. Biophysical Modeling of the Ionizing Radiation Influence on Cells Using the Stochastic (Monte Carlo) and Deterministic (Analytical) Approaches. Dose Response 2022; 20:15593258221138506. [PMID: 36458282 PMCID: PMC9706082 DOI: 10.1177/15593258221138506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/26/2022] [Indexed: 09/10/2024] Open
Abstract
This review article describes our simplified biophysical model for the response of a group of cells to ionizing radiation. The model, which is a product of 10 years of studies, acts as (a) a comprehensive stochastic approach based on the Monte Carlo simulation with a probability tree and (b) the thereof derived detailed deterministic models describing the selected biophysical and radiobiological phenomena in an analytical manner. Specifically, the presented model describes effects such as the risk of neoplastic transformation of cells relative to the absorbed radiation dose, the dynamics of tumor development, the priming dose effect (also called the Raper-Yonezawa effect) based on the introduced adaptive response approach, and the bystander effect. The model is also modifiable depending on users' potential needs.
Collapse
Affiliation(s)
- Krzysztof W. Fornalski
- Faculty of Physics, Warsaw University
of Technology (WF PW), Poland
- National Centre for Nuclear
Research (NCBJ), Poland
| | | | - Ernest Bugała
- Faculty of Physics, Warsaw University
of Technology (WF PW), Poland
| | | | | | - Jakub Kopyciński
- Center for Theoretical
Physics, Polish Academy of Sciences (CFT
PAN), Poland
| | | | - Piotr Kukulski
- Department of Mechanical, Aerospace
and Civil Engineering, University of Manchester (MACE
UoM), United Kingdom
| | | | - Julia Ponikowska
- Faculty of Physics, Warsaw University
of Technology (WF PW), Poland
| | - Joanna Reszczyńska
- Mossakowski Medical Research
Institute,
Polish Academy
of Sciences (IMDiK PAN), Poland
| | - Iwona Słonecka
- Faculty of Physics, Warsaw University
of Technology (WF PW), Poland
| | - Paweł Wysocki
- Faculty of Physics, Warsaw University
of Technology (WF PW), Poland
| | | |
Collapse
|
7
|
Peng Z, Xiong J, Dong H. Valproic Acid Inhibits Peripheral T Cell Lymphoma Cells Behaviors via Restraining PI3K/AKT Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:7350489. [PMID: 35966721 PMCID: PMC9374556 DOI: 10.1155/2022/7350489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 11/18/2022]
Abstract
Objective Alproic acid (VPA) is a clinic antiepileptic drug. Antitumor role of VPA has been studied. The aim of this study was to clarify the treatment effect and potential mechanism of VPA on peripheral T cell lymphomas (PTCLs). Materials and Methods Hut 78 cells were obtained from the Shanghai Cell Bank, Chinese Academy of Sciences, and randomly divided into six groups: control, VPA (8 mM), empty vector (NC), miR-3196 mimics, miR-3196 inhibitor, and VPA + miR-3196 mimics groups. CCK-8 assay was performed to clarify the regulative role of VPA on cell proliferation. Flow cytometry was applied to determine the apoptotic rate and ROS levels. miR-3196 was tested by RT-qPCR. Western blot was used to test the level of p-PI3K and p-AKT. Biochemical experiments were used to detect changes in the content of ATP, lactate level, and glucose content. Electron microscopy was used to show the structure of mitochondria in Hut 78 cells. Results VPA greatly promoted the expression of miR-3196 and inhibited cell proliferation in a dose-dependent manner. Compared with the NC group, the cell apoptosis rate, Bax and cleaved-caspase-3 expression, lactate level, ROS expression, and glucose content in the VPA group were significantly increased (P < 0.05), and cell proliferation, ATP production, and the expression of Bcl-2, p-PI3K and p-AKT was decreased significantly (P < 0.05). The role of mir-3196 mimics is similar to VPA. While, the miR-3196 inhibitor had the opposite effect to VPA and mimics. The combination of VPA and miR-3196 mimics has the most obvious effect. Conclusion VPA can inhibit the proliferation of Hut 78 cells and promote cell apoptosis and the structure and dysfunction of mitochondria by regulating the activity of the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Zhiqiang Peng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
- Department of Lymphatic Hematology and Oncology, Jiangxi Cancer Hospital, Nanchang 330029, Jiangxi, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Hanzhi Dong
- General Department of Oncology, Jiangxi Cancer Hospital, Nanchang 330029, Jiangxi, China
| |
Collapse
|
8
|
Ivanisenko NV, Seyrek K, Hillert-Richter LK, König C, Espe J, Bose K, Lavrik IN. Regulation of extrinsic apoptotic signaling by c-FLIP: towards targeting cancer networks. Trends Cancer 2021; 8:190-209. [PMID: 34973957 DOI: 10.1016/j.trecan.2021.12.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023]
Abstract
The extrinsic pathway is mediated by death receptors (DRs), including CD95 (APO-1/Fas) or TRAILR-1/2. Defects in apoptosis regulation lead to cancer and other malignancies. The master regulator of the DR networks is the cellular FLICE inhibitory protein (c-FLIP). In addition to its key role in apoptosis, c-FLIP may exert other cellular functions, including control of necroptosis, pyroptosis, nuclear factor κB (NF-κB) activation, and tumorigenesis. To gain further insight into the molecular mechanisms of c-FLIP action in cancer networks, we focus on the structure, isoforms, interactions, and post-translational modifications of c-FLIP. We also discuss various avenues to target c-FLIP in cancer cells for therapeutic benefit.
Collapse
Affiliation(s)
- Nikita V Ivanisenko
- The Federal Research Center Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia; Artificial Intelligence Research Institute, Moscow, Russia
| | - Kamil Seyrek
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Laura K Hillert-Richter
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Corinna König
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Johannes Espe
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Kakoli Bose
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute, BARC Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Inna N Lavrik
- The Federal Research Center Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia; Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany.
| |
Collapse
|
9
|
Emerging Therapeutic Landscape of Peripheral T-Cell Lymphomas Based on Advances in Biology: Current Status and Future Directions. Cancers (Basel) 2021; 13:cancers13225627. [PMID: 34830782 PMCID: PMC8616039 DOI: 10.3390/cancers13225627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Peripheral T-cell lymphoma is a rare but aggressive tumor. Due to its rarity, the disease has not been completely understood. In our review, we look at this lymphoma at the molecular level based on available literature. We highlight the mechanism behind the progression and resistance of this tumor. In doing so, we bring forth possible mechanism that could be exploited through novel chemotherapy drugs. In addition, we also look at the current available drugs used in treating this disease, as well as highlight other new drugs, describing their potential in treating this lymphoma. We comprehensively have collected and present the available biology behind peripheral T-cell lymphoma and discuss the available treatment options. Abstract T-cell lymphomas are a relatively rare group of malignancies with a diverse range of pathologic features and clinical behaviors. Recent molecular studies have revealed a wide array of different mechanisms that drive the development of these malignancies and may be associated with resistance to therapies. Although widely accepted chemotherapeutic agents and combinations, including stem cell transplantation, obtain responses as initial therapy for these diseases, most patients will develop a relapse, and the median survival is only 5 years. Most patients with relapsed disease succumb within 2 to 3 years. Since 2006, the USFDA has approved five medications for treatment of these diseases, and only anti-CD30-therapy has made a change in these statistics. Clearly, newer agents are needed for treatment of these disorders, and investigators have proposed studies that evaluate agents that target these malignancies and the microenvironment depending upon the molecular mechanisms thought to underlie their pathogenesis. In this review, we discuss the currently known molecular mechanisms driving the development and persistence of these cancers and discuss novel targets for therapy of these diseases and agents that may improve outcomes for these patients.
Collapse
|
10
|
Nalbantoglu S, Karadag A. Metabolomics bridging proteomics along metabolites/oncometabolites and protein modifications: Paving the way toward integrative multiomics. J Pharm Biomed Anal 2021; 199:114031. [PMID: 33857836 DOI: 10.1016/j.jpba.2021.114031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 03/02/2021] [Accepted: 03/16/2021] [Indexed: 02/08/2023]
Abstract
Systems biology adopted functional and integrative multiomics approaches enable to discover the whole set of interacting regulatory components such as genes, transcripts, proteins, metabolites, and metabolite dependent protein modifications. This interactome build up the midpoint of protein-protein/PTM, protein-DNA/RNA, and protein-metabolite network in a cell. As the key drivers in cellular metabolism, metabolites are precursors and regulators of protein post-translational modifications [PTMs] that affect protein diversity and functionality. The precisely orchestrated core pattern of metabolic networks refer to paradigm 'metabolites regulate PTMs, PTMs regulate enzymes, and enzymes modulate metabolites' through a multitude of feedback and feed-forward pathway loops. The concept represents a flawless PTM-metabolite-enzyme(protein) regulomics underlined in reprogramming cancer metabolism. Immense interconnectivity of those biomolecules in their spectacular network of intertwined metabolic pathways makes integrated proteomics and metabolomics an excellent opportunity, and the central component of integrative multiomics framework. It will therefore be of significant interest to integrate global proteome and PTM-based proteomics with metabolomics to achieve disease related altered levels of those molecules. Thereby, present update aims to highlight role and analysis of interacting metabolites/oncometabolites, and metabolite-regulated PTMs loop which may function as translational monitoring biomarkers along the reprogramming continuum of oncometabolism.
Collapse
Affiliation(s)
- Sinem Nalbantoglu
- TUBITAK Marmara Research Center, Gene Engineering and Biotechnology Institute, Molecular, Oncology Laboratory, Gebze, Kocaeli, Turkey.
| | - Abdullah Karadag
- TUBITAK Marmara Research Center, Gene Engineering and Biotechnology Institute, Molecular, Oncology Laboratory, Gebze, Kocaeli, Turkey
| |
Collapse
|
11
|
Basak D, Uddin MN, Hancock J. The Role of Oxidative Stress and Its Counteractive Utility in Colorectal Cancer (CRC). Cancers (Basel) 2020; 12:E3336. [PMID: 33187272 PMCID: PMC7698080 DOI: 10.3390/cancers12113336] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022] Open
Abstract
An altered redox status accompanied by an elevated generation of reactive oxygen/nitrogen species (ROS/RNS) has been implicated in a number of diseases including colorectal cancer (CRC). CRC, being one of the most common cancers worldwide, has been reported to be associated with multiple environmental and lifestyle factors (e.g., dietary habits, obesity, and physical inactivity) and harboring heightened oxidative stress that results in genomic instability. Although under normal condition ROS regulate many signal transduction pathways including cell proliferation and survival, overwhelming of the antioxidant capacity due to metabolic abnormalities and oncogenic signaling leads to a redox adaptation response that imparts drug resistance. Nevertheless, excessive reliance on elevated production of ROS makes the tumor cells increasingly vulnerable to further ROS insults, and the abolition of such drug resistance through redox perturbation could be instrumental to preferentially eliminate them. The goal of this review is to demonstrate the evidence that links redox stress to the development of CRC and assimilate the most up-to-date information that would facilitate future investigation on CRC-associated redox biology. Concomitantly, we argue that the exploitation of this distinct biochemical property of CRC cells might offer a fresh avenue to effectively eradicate these cells.
Collapse
Affiliation(s)
- Debasish Basak
- College of Pharmacy, Larkin University, Miami, FL 33169, USA;
| | | | - Jake Hancock
- College of Pharmacy, Larkin University, Miami, FL 33169, USA;
| |
Collapse
|
12
|
Zhang P, Zhang M. Epigenetic alterations and advancement of treatment in peripheral T-cell lymphoma. Clin Epigenetics 2020; 12:169. [PMID: 33160401 PMCID: PMC7648940 DOI: 10.1186/s13148-020-00962-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 10/28/2020] [Indexed: 02/08/2023] Open
Abstract
Peripheral T-cell lymphoma (PTCL) is a rare and heterogeneous group of clinically aggressive diseases associated with poor prognosis. Except for ALK + anaplastic large-cell lymphoma (ALCL), most peripheral T-cell lymphomas are highly malignant and have an aggressive disease course and poor clinical outcomes, with a poor remission rate and frequent relapse after first-line treatment. Aberrant epigenetic alterations play an important role in the pathogenesis and development of specific types of peripheral T-cell lymphoma, including the regulation of the expression of genes and signal transduction. The most common epigenetic alterations are DNA methylation and histone modification. Histone modification alters the level of gene expression by regulating the acetylation status of lysine residues on the promoter surrounding histones, often leading to the silencing of tumour suppressor genes or the overexpression of proto-oncogenes in lymphoma. DNA methylation refers to CpG islands, generally leading to tumour suppressor gene transcriptional silencing. Genetic studies have also shown that some recurrent mutations in genes involved in the epigenetic machinery, including TET2, IDH2-R172, DNMT3A, RHOA, CD28, IDH2, TET2, MLL2, KMT2A, KDM6A, CREBBP, and EP300, have been observed in cases of PTCL. The aberrant expression of miRNAs has also gradually become a diagnostic biomarker. These provide a reasonable molecular mechanism for epigenetic modifying drugs in the treatment of PTCL. As epigenetic drugs implicated in lymphoma have been continually reported in recent years, many new ideas for the diagnosis, treatment, and prognosis of PTCL originate from epigenetics in recent years. Novel epigenetic-targeted drugs have shown good tolerance and therapeutic effects in the treatment of peripheral T-cell lymphoma as monotherapy or combination therapy. NCCN Clinical Practice Guidelines also recommended epigenetic drugs for PTCL subtypes as second-line therapy. Epigenetic mechanisms provide new directions and therapeutic strategies for the research and treatment of peripheral T-cell lymphoma. Therefore, this paper mainly reviews the epigenetic changes in the pathogenesis of peripheral T-cell lymphoma and the advancement of epigenetic-targeted drugs in the treatment of peripheral T-cell lymphoma (PTCL).
Collapse
Affiliation(s)
- Ping Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan Province, China.,Academy of Medical Sciences of Zhengzhou University, Zhengzhou City, 450052, Henan Province, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan Province, China. .,Academy of Medical Sciences of Zhengzhou University, Zhengzhou City, 450052, Henan Province, China.
| |
Collapse
|
13
|
Surapally S, Jayaprakasam M, Verma RS. Curcumin augments therapeutic efficacy of TRAIL-based immunotoxins in leukemia. Pharmacol Rep 2020; 72:1032-1046. [PMID: 32141025 DOI: 10.1007/s43440-020-00073-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/26/2019] [Accepted: 12/30/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo2L) has been perceived as a promising anti-cancer agent because of its unique ability to kill cancer cells while sparing normal cells. However, translation of TRAIL to clinical studies was less successful as a large number of cancer cells acquire resistance to TRAIL-based monotherapies. An ideal strategy to overcome TRAIL resistance is to combine it with potential sensitizing agents. OBJECTIVE To investigate the TRAIL-sensitizing effect of curcumin in leukemia. METHODS The mechanism underlying TRAIL sensitization by curcumin was studied by flow cytometric analysis of TRAIL receptors in leukemic cell lines and patient samples, and immunoblot detection of TRAIL-apoptosis signaling proteins. RESULTS Curcumin augments TRAIL-apoptotic signaling in leukemic cells by upregulating the expression of DR4 and DR5 along with suppression of cFLIP and anti-apoptotic proteins Mcl-1, Bcl-xl, and XIAP. Curcumin pre-treatment significantly (p < 0.01) enhanced the sensitivity of leukemic cell lines to TRAIL recombinant proteins. IL2-TRAIL peptide in the presence of curcumin induced potent apoptosis (p < 0.001) as compared to TRAIL and IL2-TRAIL protein in leukemic cell lines with IC50 < 0.1 μΜ. Additionally, the combination of IL2-TRAIL peptide and curcumin showed significant cytotoxicity in patient peripheral blood mononuclear cells (PBMCs) with an efficacy of 90% in acute myeloid leukemia (AML), but 100% in acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL) and chronic myelomonocytic leukemia (CMML). CONCLUSION Overall, our results suggest that curcumin potentiates TRAIL-induced apoptosis through modulation of death receptors and anti-apoptotic proteins which significantly enhances the therapeutic efficacy.
Collapse
Affiliation(s)
- Sridevi Surapally
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences Building, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India
| | - Madhumathi Jayaprakasam
- Division of Epidemiology and Communicable Diseases, Indian Council for Medical Research (ICMR), New Delhi, India
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences Building, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India.
| |
Collapse
|
14
|
Chi Z, Gao H, Liu H, Wu B, Zhang B, Gu M, Yang W. Chidamide induces necroptosis via regulation of c‑FLIPL expression in Jurkat and HUT‑78 cells. Mol Med Rep 2019; 21:936-944. [PMID: 31974619 DOI: 10.3892/mmr.2019.10873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/14/2019] [Indexed: 11/06/2022] Open
Abstract
T‑cell acute lymphoblastic leukemia (T‑ALL) is a hematopoietic malignancy, which is associated with a poor prognosis. It is difficult to achieve complete remission or long‑term survival with conventional chemotherapy, partly due to decreased apoptosis. However, necroptosis can serve as an alternative pathway to induce cell death. The present study investigated whether the selective histone deacetylase (HDAC) inhibitor chidamide exerted a therapeutic effect on T‑ALL and explored the underlying mechanism. The results revealed that HDAC expression was increased in Jurkat and HUT‑78 cells treated compared with the control cell line (H9), and was accompanied by elevated cellular Fas‑associated death domain‑like interleukin‑1β converting enzyme inhibitory protein long form (c‑FLIPL) levels. Chidamide treatment (2 µmol/l) also induced mitochondrial dysfunction, necroptosis and apoptosis in T‑ALL cells in vitro. Furthermore, necroptosis was increased when apoptosis was blocked in T‑ALL cells. Additionally, chidamide (2 µmol/l) downregulated c‑FLIPL, HDAC1 and HDAC3 expression, and increased receptor‑interacting protein kinase 3 expression and the phosphorylation of mixed lineage kinase domain‑like pseudokinase in Jurkat and HUT‑78 cells. The results obtained in the present study revealed that chidamide may induce necroptosis via regulation of c‑FLIPL expression when apoptosis is inhibited in Jurkat and HUT‑78 cells.
Collapse
Affiliation(s)
- Zuofei Chi
- Department of Pediatric Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Hongyu Gao
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Hui Liu
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Bin Wu
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Bin Zhang
- Department of Pediatric Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Min Gu
- Department of Pediatric Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Wei Yang
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| |
Collapse
|
15
|
Lee BS, Kim YS, Kim HJ, Kim DH, Won HR, Kim YS, Kim CH. HDAC4 degradation by combined TRAIL and valproic acid treatment induces apoptotic cell death of TRAIL-resistant head and neck cancer cells. Sci Rep 2018; 8:12520. [PMID: 30131570 PMCID: PMC6104079 DOI: 10.1038/s41598-018-31039-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 08/03/2018] [Indexed: 01/30/2023] Open
Abstract
Although TRAIL can directly induce cell death in some cancer cells, it appears that TRAIL resistance exists in many cancers. This study focuses on anti-cancer drugs for TRAIL-resistant head and neck cancer (HNC) to provide further progress toward effective cancer therapy. Results indicate in TRAIL-resistant HNC cells, that combined TRAIL and VPA treatment greatly reduced cell viability and therefore induced cell death, relative to treatment with TRAIL or VPA alone. A caspase-dependent signaling pathway was demonstrated, and combined treatment with TRAIL and VPA also significantly decreased the expression of HDAC4. When we pretreated cells with z-VAD followed by combined treatment with TRAIL and VPA, cell death was blocked with no reduction in expression of HDAC4. To confirm that cell death involved HDAC4 in HNC cells, we knocked down expression of HDAC4 with siRNA, followed by treatment with TRAIL and VPA. Results showed that loss of HDAC4 sensitized the TRAIL-resistant HNC cells to apoptotic cell death. Finally, we showed elevated expression of HDAC4 in HNC tissues compared to normal tissues obtained from the same patients. In conclusion, we suggest that combined VPA and TRAIL treatment may be a promising therapy for HNC via HDAC4 degradation.
Collapse
Affiliation(s)
- Bok-Soon Lee
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Yeon Soo Kim
- Department of Otorhinolaryngology, College of Medicine, Konyang University Hospital, Konyang University Myunggok Medical Research Institute, Daejeon, 35365, Republic of Korea
| | - Haeng-Jun Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea.,Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Dae-Ho Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea.,Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Ho-Ryun Won
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea.,Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University, Daejeon, 35015, Republic of Korea
| | - Yong-Sung Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Chul-Ho Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea. .,Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea.
| |
Collapse
|
16
|
Multiple Functions of Cellular FLIP Are Essential for Replication of Hepatitis B Virus. J Virol 2018; 92:JVI.00339-18. [PMID: 29875248 DOI: 10.1128/jvi.00339-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/24/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) infection is a leading cause of liver diseases; however, the host factors which facilitate the replication and persistence of HBV are largely unidentified. Cellular FLICE inhibitory protein (c-FLIP) is a typical antiapoptotic protein. In many cases of liver diseases, the expression level of c-FLIP is altered, which affects the fate of hepatocytes. We previously found that c-FLIP and its cleaved form interact with HBV X protein (HBx), which is essential for HBV replication, and regulate diverse cellular signals. In this study, we investigated the role of endogenous c-FLIP in HBV replication and its underlying mechanisms. The knockdown of endogenous c-FLIP revealed that this protein regulates HBV replication through two different mechanisms. (i) c-FLIP interacts with HBx and protects it from ubiquitin-dependent degradation. The N-terminal DED1 domain of c-FLIP is required for HBx stabilization. (ii) c-FLIP regulates the expression or stability of hepatocyte nuclear factors (HNFs), which have critical roles in HBV transcription and maintenance of hepatocytes. c-FLIP regulates the stability of HNFs through physical interactions. We verified our findings in three HBV infection systems: HepG2-NTCP cells, differentiated HepaRG cells, and primary human hepatocytes. In conclusion, our results identify c-FLIP as an essential factor in HBV replication. c-FLIP regulates viral replication through its multiple effects on viral and host proteins that have critical roles in HBV replication.IMPORTANCE Although the chronic hepatitis B virus (HBV) infection still poses a major health concern, the host factors which are required for the replication of HBV are largely uncharacterized. Our studies identify cellular FLICE inhibitory protein (c-FLIP) as an essential factor in HBV replication. We found the dual roles of c-FLIP in regulation of HBV replication: c-FLIP interacts with HBx and enhances its stability and regulates the expression or stability of hepatocyte nuclear factors which are essential for transcription of HBV genome. Our findings may provide a new target for intervention in persistent HBV infection.
Collapse
|
17
|
Huang Y, Yang W, Zeng H, Hu C, Zhang Y, Ding N, Fan G, Shao L, Kuang B. Droxinostat sensitizes human colon cancer cells to apoptotic cell death via induction of oxidative stress. Cell Mol Biol Lett 2018; 23:34. [PMID: 30065760 PMCID: PMC6064062 DOI: 10.1186/s11658-018-0101-5] [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: 01/04/2018] [Accepted: 07/19/2018] [Indexed: 01/08/2023] Open
Abstract
Upregulation of histone acetylation plays a critical role in the dysregulation of transcription. It alters the structure of chromatin, which leads to the onset of cancer. Histone deacetylase inhibitors may therefore be a promising way to limit cancer progression. In this study, we examined the effects of droxinostat on the growth of HT-29 colon cancer cells. Our results show that droxinostat effectively inhibited cell growth and colony-forming ability by inducing cellular apoptosis and ROS production in HT-29 cells. Notably, the apoptotic inhibitor Z-VAD-FMK significantly decreased the levels of cellular apoptosis and the antioxidant γ-tocotrienol (GT3) significantly decreased ROS production induced by droxinostat treatment. Z-VAD-FMK and GT3 also partially reversed the negative growth effects of droxinstat on HT-29 cells. GT3 treatment decreased cellular apoptosis and increased colony-forming ability upon droxinostat administration. Z-VAD-FMK treatment also partially decreased droxinostat-induced ROS production. Our findings suggest that the effects of droxinostat on colon cancer cells are mediated by the induction of oxidative stress and apoptotic cell death.
Collapse
Affiliation(s)
- Ying Huang
- 1Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006 China.,2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| | - Wuping Yang
- 1Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006 China.,2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| | - Huihong Zeng
- 2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| | - Chuan Hu
- 2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| | - Yaqiong Zhang
- 2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| | - Nanhua Ding
- 2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| | - Guangqin Fan
- 1Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006 China.,3School of Public Health, Nanchang University, Nanchang, 330006 China
| | - Lijian Shao
- 1Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006 China.,3School of Public Health, Nanchang University, Nanchang, 330006 China
| | - Bohai Kuang
- 2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| |
Collapse
|
18
|
Pizzi M, Margolskee E, Inghirami G. Pathogenesis of Peripheral T Cell Lymphoma. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 13:293-320. [DOI: 10.1146/annurev-pathol-020117-043821] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Marco Pizzi
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY 10021, USA
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padova, 35121 Padova, Italy
| | - Elizabeth Margolskee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY 10021, USA
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY 10021, USA
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies (CeRMS), University of Torino, 10126 Torino, Italy
- Department of Pathology and NYU Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| |
Collapse
|
19
|
Rezk Hassan GF, Marey K. Immunohistopathological Study of c-FLIP Protein in Mycosis Fungoides. Asian Pac J Cancer Prev 2017; 18:2493-2499. [PMID: 28952283 PMCID: PMC5720656 DOI: 10.22034/apjcp.2017.18.9.2493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Mycosis fungoides (MF) is the commonest variant of primary cutaneous T cell lymphoma with several clinicopathologic variants. Defective apoptotic mechanism may be important in the pathogenesis and progression of MF. c-FLIP protein is an important anti-apoptotic marker and chemotherapeutic resistant factor. This study aimed to evaluate the c-FLIP expression in MF and its role in the pathogenesis of MF. Methods: Twenty patients of MF and ten normal persons were included in this study. Skin biopsies were obtained from both patients and controls. They were studied and examined immunohistochemically for the expression of CD4 and c-FLIP. Results: c-FLIP expression was significantly increased in patients when compared to controls in both epidermis and dermis. There were positive correlations between c-FLIP expression and CD4+ expression in both epidermal and dermal lesions of patients group. There were statistically significant positive correlations between c-FLIP expression (in both dermal and epidermal lesions) and the age of patients. c-FLIP expression increased with the tumor progression but with no statistical significance. Conclusion: Defective regulation of apoptosis has been considered as a main cause for accumulation of clonal T cells, and it was related to an increased expression of c-FLIP which may have a role in the pathogenesis of MF. Also, c-FLIP may have prognostic information in MF as its level increased with both age of the patients and tumor progression.
Collapse
Affiliation(s)
- Ghada Fawzy Rezk Hassan
- Lecturer of Dermatology and Venereology, Faculty of medicine, Tanta University, 31111, Tanta, Egypt.
| | | |
Collapse
|
20
|
Xu PP, Sun YF, Fang Y, Song Q, Yan ZX, Chen Y, Jiang XF, Fei XC, Zhao Y, Leboeuf C, Li B, Wang CF, Janin A, Wang L, Zhao WL. JAM-A overexpression is related to disease progression in diffuse large B-cell lymphoma and downregulated by lenalidomide. Sci Rep 2017; 7:7433. [PMID: 28785100 PMCID: PMC5547054 DOI: 10.1038/s41598-017-07964-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/06/2017] [Indexed: 01/10/2023] Open
Abstract
Cancer stem cells play an important role on tumor progression. Biomarkers of stem cell property and their relationship to extranodal involvement of malignant lymphocytes are undefined in diffuse large B-cell lymphoma (DLBCL). Here we showed that junctional adhesion molecule-A (JAM-A) was highly expressed in DLBCL patients with multiple extranodal lesions. JAM-A maintained B-lymphoma cell stemness and was associated with cell invasion and epithelial-to-mesenchymal transition both in vitro and in vivo. As mechanism of action, JAM-A overexpression selectively activated transforming growth factor-β (TGF-β)/NODAL signaling, thereby enhanced B-lymphoma cell aggressiveness and induced extranodal involvement to mesoendoderm-derived organs in DLBCL. Lenalidomide downregulated JAM-A and downstream NODAL expression, resulting in inhibition of B-lymphoma cell invasion and epithelial-to-mesenchymal transition. In a murine xenograft model established with subcutaneous injection of JAM-A-overexpressing B-lymphoma cells, lenalidomide retarded tumor growth and prevented cell invasion to mesoendoderm-derived organs, consistent with the downregulation of JAM-A and NODAL expression. Collectively, these findings indicated that JAM-A was related to extranodal involvement in DLBCL through modulating TGF-β/NODAL signaling. Identified as a biomarker of stem cell property, JAM-A indicated the sensitivity of B-lymphoma cells to lenalidomide. Therapeutic targeting of JAM-A/NODAL axis could thus be a promising clinical strategy to impede tumor progression in DLBCL.
Collapse
Affiliation(s)
- Peng-Peng Xu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France
| | - Yi-Feng Sun
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France
| | - Ying Fang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France
| | - Qi Song
- Department of Radiology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France
| | - Zi-Xun Yan
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France
| | - Yi Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France
| | - Xu-Feng Jiang
- Department of Nuclear Medicine, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France
| | - Xiao-Chun Fei
- Department of Pathology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France
| | - Yan Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France
| | - Christophe Leboeuf
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France.,U1165 Inserm/Université Paris 7, Hôpital Saint Louis, Pairs, France
| | - Biao Li
- Department of Nuclear Medicine, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France
| | - Chao-Fu Wang
- Department of Pathology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France
| | - Anne Janin
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France.,U1165 Inserm/Université Paris 7, Hôpital Saint Louis, Pairs, France
| | - Li Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France. .,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France.
| | - Wei-Li Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France. .,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Pairs, France.
| |
Collapse
|
21
|
Xiong J, Wang L, Fei XC, Jiang XF, Zheng Z, Zhao Y, Wang CF, Li B, Chen SJ, Janin A, Gale RP, Zhao WL. MYC is a positive regulator of choline metabolism and impedes mitophagy-dependent necroptosis in diffuse large B-cell lymphoma. Blood Cancer J 2017; 7:e0. [PMID: 28686226 PMCID: PMC5549253 DOI: 10.1038/bcj.2017.61] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/19/2017] [Accepted: 06/01/2017] [Indexed: 02/07/2023] Open
Abstract
The activation of oncogenes can reprogram tumor cell metabolism. Here, in diffuse large B-cell lymphoma (DLBCL), serum metabolomic analysis revealed that oncogenic MYC could induce aberrant choline metabolism by transcriptionally activating the key enzyme phosphate cytidylyltransferase 1 choline-α (PCYT1A). In B-lymphoma cells, as a consequence of PCYT1A upregulation, MYC impeded lymphoma cells undergo a mitophagy-dependent necroptosis. In DLBCL patients, overexpression of PCYT1A was in parallel with an increase in tumor MYC, as well as a decrease in serum choline metabolite phosphatidylcholine levels and an International Prognostic Index, indicating intermediate-high or high risk. Both in vitro and in vivo, lipid-lowering alkaloid berberine (BBR) exhibited an anti-lymphoma activity through inhibiting MYC-driven downstream PCYT1A expression and inducing mitophagy-dependent necroptosis. Collectively, PCYT1A was upregulated by MYC, which resulted in the induction of aberrant choline metabolism and the inhibition of B-lymphoma cell necroptosis. Referred as a biomarker for DLBCL progression, PCYT1A can be targeted by BBR, providing a potential lipid-modifying strategy in treating MYC-High lymphoma.
Collapse
Affiliation(s)
- J Xiong
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai Rui Jin Hospital, Shanghai, China
| | - L Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai Rui Jin Hospital, Shanghai, China
| | - X-C Fei
- Department of Pathology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - X-F Jiang
- Department of Nuclear Medicine, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Z Zheng
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai Rui Jin Hospital, Shanghai, China
| | - Y Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai Rui Jin Hospital, Shanghai, China
| | - C-F Wang
- Department of Pathology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - B Li
- Department of Nuclear Medicine, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - S-J Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai Rui Jin Hospital, Shanghai, China
| | - A Janin
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai Rui Jin Hospital, Shanghai, China.,Laboratory of Pathology, Paris Diderot University, U1165 Inserm, Paris, France
| | - R P Gale
- Haematology Research Centre, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, UK
| | - W-L Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai Rui Jin Hospital, Shanghai, China
| |
Collapse
|
22
|
Zheng Z, Xu PP, Wang L, Zhao HJ, Weng XQ, Zhong HJ, Qu B, Xiong J, Zhao Y, Wang XF, Janin A, Zhao WL. MiR21 sensitized B-lymphoma cells to ABT-199 via ICOS/ICOSL-mediated interaction of Treg cells with endothelial cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017. [PMID: 28637496 PMCID: PMC5480196 DOI: 10.1186/s13046-017-0551-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND MicroRNAs (miRs) are involved in tumor progression by regulating tumor cells and tumor microenvironment. MiR21 is overexpressed in diffuse large B-cell lymphoma (DLBCL) and its biological impact on tumor microenvironment remains unclear. METHODS MiR21 was assessed by quantitative RT-PCR in patients with newly diagnosed DLBCL. The mechanism of action of miR21 on lymphoma progression and tumor angiogenesis was examined in vitro in B-lymphoma cell lines and in vivo in a murine xenograft model. RESULTS Serum miR21 was significantly elevated in patients and associated with advanced disease stage, International Prognostic Index indicating intermediate-high and high-risk, and increased tumor angiogenesis. When co-cultured with immune cells and endothelial cells, miR21-overexpressing B-lymphoma cells were resistant to chemotherapeutic agents, but sensitive to Bcl-2 inhibitor ABT-199, irrespective of Bcl-2 expression on lymphoma cells. In both co-culture systems of Bcl-2positive and Bcl-2negative B-lymphoma cells, miR21 induced inducible co-stimulator (ICOS) expression on regulatory T (Treg) cells. Through crosstalking with Treg cells by ICOS ligand (ICOSL), endothelial cells were activated, resulting in stimulation of Bcl-2 expression and vessel formation. ABT-199 directly targeted Bcl-2 on endothelial cells, induced endothelial cell apoptosis and inhibited tumor angiogenesis. In a murine xenograft model established with subcutaneous injection of B-lymphoma cells, ABT-199 particularly retarded the growth of miR21-overexpressing tumors, consistent with the induction of endothelial cell apoptosis and inhibition of tumor angiogenesis. CONCLUSIONS As a serum oncogenic biomarker of B-cell lymphoma, miR21 indicated B-lymphoma cell sensitivity to ABT-199 via ICOS/ICOSL-mediated interaction of Treg cells with endothelial cells.
Collapse
Affiliation(s)
- Zhong Zheng
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Peng-Peng Xu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China.,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China
| | - Li Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China.,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China
| | - Hui-Jin Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Xiang-Qin Weng
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Hui-Juan Zhong
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Bin Qu
- Department of Laboratory Medicine, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Xiong
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Yan Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Xue-Feng Wang
- Department of Laboratory Medicine, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Anne Janin
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China.,U1165 Inserm/Université Paris 7, Hôpital Saint Louis, Paris, France
| | - Wei-Li Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China. .,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China.
| |
Collapse
|
23
|
Zhou L, Zheng H, Huang X, Zhu L, Wu S, Zeng C, Yang L, Chen S, Luo G, Du X, Li Y. Different genetic alteration of A20
in a Sézary syndrome case with Vα2-Jα22
T cell clone. Asia Pac J Clin Oncol 2017; 14:e116-e123. [PMID: 28296250 DOI: 10.1111/ajco.12672] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/13/2017] [Indexed: 01/28/2023]
Affiliation(s)
- Lingling Zhou
- Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
- Institute of Hematology, School of Medicine; Jinan University; Guangzhou China
| | - Haitao Zheng
- Institute of Hematology, School of Medicine; Jinan University; Guangzhou China
| | - Xin Huang
- Department of Hematology; Guangdong General Hospital (Guangdong Academy of Medical Sciences); Guangzhou China
| | - Lihua Zhu
- Department of Rheumatism and Immunology; First Affiliated Hospital; Jinan University; Guangzhou China
| | - Suijing Wu
- Department of Hematology; Guangdong General Hospital (Guangdong Academy of Medical Sciences); Guangzhou China
| | - Chengwu Zeng
- Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
- Institute of Hematology, School of Medicine; Jinan University; Guangzhou China
| | - Lijian Yang
- Institute of Hematology, School of Medicine; Jinan University; Guangzhou China
| | - Shaohua Chen
- Institute of Hematology, School of Medicine; Jinan University; Guangzhou China
| | - Gengxin Luo
- Department of Hematology; First Affiliated Hospital; Jinan University; Guangzhou China
| | - Xin Du
- Department of Hematology; Guangdong General Hospital (Guangdong Academy of Medical Sciences); Guangzhou China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
- Institute of Hematology, School of Medicine; Jinan University; Guangzhou China
- Department of Hematology; First Affiliated Hospital; Jinan University; Guangzhou China
| |
Collapse
|
24
|
Lv XQ, Qiao XR, Su L, Chen SZ. Honokiol inhibits EMT-mediated motility and migration of human non-small cell lung cancer cells in vitro by targeting c-FLIP. Acta Pharmacol Sin 2016; 37:1574-1586. [PMID: 27593221 PMCID: PMC5290996 DOI: 10.1038/aps.2016.81] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/12/2016] [Indexed: 12/12/2022]
Abstract
AIM Honokiol (HNK) is a natural compound isolated from the magnolia plant with numerous pharmacological activities, including inhibiting epithelial-mesenchymal transition (EMT), which has been proposed as an attractive target for anti-tumor drugs to prevent tumor migration. In this study we investigated the effects of HNK on EMT in human NSCLC cells in vitro and the related signaling mechanisms. METHODS TNF-α (25 ng/mL) in combination with TGF-β1 (5 ng/mL) was used to stimulate EMT of human NSCLC A549 and H460 cells. Cell proliferation was analyzed using a sulforhodamine B assay. A wound-healing assay and a transwell assay were performed to examine cell motility. Western blotting was used to detect the expression levels of relevant proteins. siRNAs were used to knock down the gene expression of c-FLIP and N-cadherin. Stable overexpression of c-FLIP L (H157-FLIP L) or Lac Z (H157-Lac Z) was also performed. RESULTS Treatment with TNF-α+TGF-β1 significantly enhanced the migration of A549 and H460 cells, increased c-FLIP, N-cadherin (a mesenchymal marker), snail (a transcriptional modulator) and p-Smad2/3 expression, and decreased IκB levels in the cells; these changes were abrogated by co-treatment with HNK (30 μmol/L). Further studies demonstrated that expression level of c-FLIP was highly correlated with the movement and migration of NSCLC cells, and the downstream effectors of c-FLIP signaling were NF-κB signaling and N-cadherin/snail signaling, while Smad signaling might lie upstream of c-FLIP. CONCLUSION HNK inhibits EMT-mediated motility and migration of human NSCLC cells in vitro by targeting c-FLIP, which can be utilized as a promising target for cancer therapy, while HNK may become a potential anti-metastasis drug or lead compound.
Collapse
Affiliation(s)
- Xiao-qin Lv
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xin-ran Qiao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ling Su
- Shandong University School of Life Sciences, Ji-nan 250100, China
| | - Shu-zhen Chen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
25
|
Song Y, Wu F, Wu J. Targeting histone methylation for cancer therapy: enzymes, inhibitors, biological activity and perspectives. J Hematol Oncol 2016; 9:49. [PMID: 27316347 PMCID: PMC4912745 DOI: 10.1186/s13045-016-0279-9] [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: 04/06/2016] [Accepted: 06/07/2016] [Indexed: 12/31/2022] Open
Abstract
Post-translational methylation of histone lysine or arginine residues plays important roles in gene regulation and other physiological processes. Aberrant histone methylation caused by a gene mutation, translocation, or overexpression can often lead to initiation of a disease such as cancer. Small molecule inhibitors of such histone modifying enzymes that correct the abnormal methylation could be used as novel therapeutics for these diseases, or as chemical probes for investigation of epigenetics. Discovery and development of histone methylation modulators are in an early stage and undergo a rapid expansion in the past few years. A number of highly potent and selective compounds have been reported, together with extensive preclinical studies of their biological activity. Several compounds have been in clinical trials for safety, pharmacokinetics, and efficacy, targeting several types of cancer. This review summarizes the biochemistry, structures, and biology of cancer-relevant histone methylation modifying enzymes, small molecule inhibitors and their preclinical and clinical antitumor activities. Perspectives for targeting histone methylation for cancer therapy are also discussed.
Collapse
Affiliation(s)
- Yongcheng Song
- Department of Pharmacology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA.
| | - Fangrui Wu
- Department of Pharmacology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Jingyu Wu
- Department of Pharmacology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| |
Collapse
|
26
|
Affiliation(s)
- Katelyn Hood
- Nationwide Children's Hospital, Columbus, Ohio, and Georgia Regents Medical Center, The University of Georgia College of Pharmacy, Augusta, Georgia
| | - Arpita Shah
- Nationwide Children's Hospital, Columbus, Ohio, and Georgia Regents Medical Center, The University of Georgia College of Pharmacy, Augusta, Georgia
| |
Collapse
|
27
|
Guo ZL, Yu B, Ning BT, Chan S, Lin QB, Li JCB, Huang JD, Chan GCF. Genetically modified "obligate" anaerobic Salmonella typhimurium as a therapeutic strategy for neuroblastoma. J Hematol Oncol 2015; 8:99. [PMID: 26286454 PMCID: PMC4545364 DOI: 10.1186/s13045-015-0196-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 08/06/2015] [Indexed: 12/18/2022] Open
Abstract
Background Neuroblastoma currently has poor prognosis, therefore we proposed a new strategy by targeting neuroblastoma with genetically engineered anaerobic Salmonella (Sal-YB1). Methods Nude and nonobese diabetic-severe combined immunodeficiency (NOD-SCID) orthotopic mouse models were used, and Sal-YB1 was administered via tail vein. The therapeutic effectiveness, bio-safety, and mechanisms were studied. Results No mice died of therapy-related complications. Tumor size reduction was 70 and 30 % in nude and NOD-SCID mice, respectively. No Salmonella was detected in the urine; 75 % mice had positive stool culture if diaminopimelic acid was added, but all turned negative subsequently. Tumor tissues had more Sal-YB1 infiltration, necrosis, and shrinkage in Sal-YB1-treated mice. Significantly higher expression of TLR4, TNF-stimulated gene 6 protein (TSG6), and cleaved caspase 1, 3, 8, and 9 was found in the tumor masses of the Sal-YB1-treated group with a decrease of interleukin 1 receptor-associated kinase (IRAK) and nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha (IκBα). There was a high release of TNFα both in human macrophages and mouse tumor tissues with Sal-YB1 treatment. The antitumor effect of the supernatant derived from macrophages treated with Sal-YB1 could be reversed with TNFα and pan-caspase inhibitors. Conclusions This new approach in targeting neuroblastoma by bio-engineered Salmonella with the assistance of macrophages indirectly may have a clinical therapeutic impact in the future.
Collapse
Affiliation(s)
- Zhu-Ling Guo
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, SAR, People's Republic of China.
| | - Bin Yu
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, SAR, People's Republic of China. .,Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, People's Republic of China.
| | - Bo-Tao Ning
- Department of Hematology & Oncology of Children's Hospital, Zhejiang Key Laboratory for Diagnosis and Treatment of Neonatal Diseases, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
| | - Shing Chan
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, SAR, People's Republic of China.
| | - Qiu-Bin Lin
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, SAR, People's Republic of China.
| | - James Chun-Bong Li
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, SAR, People's Republic of China.
| | - Jian-Dong Huang
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, SAR, People's Republic of China. .,HKU-SIRI, the University of Hong Kong, Hong Kong, SAR, People's Republic of China.
| | - Godfrey Chi-Fung Chan
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, SAR, People's Republic of China.
| |
Collapse
|