1
|
Lyu H, Ishimura A, Suzuki R, Buyanbat K, Batbayar G, Meguro-Horike M, Horike SI, Yano S, Suzuki T. HDAC5, an early osimertinib-responsive gene, is a novel therapeutic target for the drug resistance in EGFR-mutant lung adenocarcinoma cells. Biochem Biophys Rep 2025; 42:102016. [PMID: 40290805 PMCID: PMC12022642 DOI: 10.1016/j.bbrep.2025.102016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
Aberrant epigenetic regulation is closely associated with drug tolerance, an early step in the acquisition of drug resistance. We previously reported that a pioneer transcriptional factor (also called an epigenetic initiator) rapidly induced by osimertinib, a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, plays a pivotal role in promoting the formation of osimertinib-tolerant cells. In this study, to identify novel epigenetic factors associated with osimertinib-tolerance, we performed a comprehensive screening of epigenetic factors whose expression is rapidly induced by osimertinib. Our results revealed that HDAC5, a class IIa histone deacetylase (HDAC), is a prominently induced epigenetic regulator in several EGFR-mutant non-small cell lung cancer (NSCLC) cell lines during the early response to osimertinib. Knockdown of HDAC5 significantly reduced the emergence of osimertinib-resistant cells. Furthermore, treatment with LMK235, a selective HDAC5 inhibitor, significantly increased global histone acetylation and enhanced osimertinib-induced apoptosis. These findings highlight the potential of HDAC5 as a novel therapeutic target to overcome osimertinib-resistance and suggest LMK235 as a promising compound to provide therapeutic benefit to EGFR-mutant NSCLC patients receiving osimertinib treatment.
Collapse
Affiliation(s)
- Hanbing Lyu
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Akihiko Ishimura
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Ryusuke Suzuki
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Khurelsukh Buyanbat
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Gerelsuren Batbayar
- Laboratory of Molecular Biology, Institute of Biology, Mongolian Academy of Sciences, Mongolia
| | - Makiko Meguro-Horike
- Division of Integrated Omics Research, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Shin-ichi Horike
- Division of Integrated Omics Research, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Seiji Yano
- Department of Respiratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical, and Health Sciences, Kanazawa University, Kanazawa, Japan
- Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Takeshi Suzuki
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
2
|
Zhang Y, Wang H, Zhan Z, Gan L, Bai O. Mechanisms of HDACs in cancer development. Front Immunol 2025; 16:1529239. [PMID: 40260239 PMCID: PMC12009879 DOI: 10.3389/fimmu.2025.1529239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
Histone deacetylases (HDACs) are a class of epigenetic regulators that play pivotal roles in key biological processes such as cell proliferation, differentiation, metabolism, and immune regulation. Based on this, HDAC inhibitors (HDACis), as novel epigenetic-targeted therapeutic agents, have demonstrated significant antitumor potential by inducing cell cycle arrest, activating apoptosis, and modulating the immune microenvironment. Current research is focused on developing highly selective HDAC isoform inhibitors and combination therapy strategies tailored to molecular subtypes, aiming to overcome off-target effects and resistance issues associated with traditional broad-spectrum inhibitors. This review systematically elaborates on the multidimensional regulatory networks of HDACs in tumor malignancy and assesses the clinical translation progress of next-generation HDACis and their prospects in precision medicine, providing a theoretical framework and strategic reference for the development of epigenetic-targeted antitumor drugs.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Haotian Wang
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Zhumei Zhan
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Lin Gan
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Out Bai
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Fang Z, Gong C, Hu Y, Cui T, Lin M, Lin S, Ye M. Acetylation of E2F1 at K125 facilitates cell apoptosis under serum stress. Transl Oncol 2025; 52:102259. [PMID: 39731810 PMCID: PMC11743828 DOI: 10.1016/j.tranon.2024.102259] [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: 07/24/2024] [Revised: 11/24/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024] Open
Abstract
E2F1 is a critical transcription factor that regulates cell cycle progression, is expressed at high levels in most cancer cells, and activates the biogenesis of proteins related to the cell cycle. Over recent years, researchers have demonstrated that E2F1 could also facilitate cellular apoptosis under conditions of cellular stress, thus creating a double-edged sword associated with both the regulation of cellular survival and death. However, the mechanisms responsible for these actions remain poorly understood. In this study, we demonstrated that serum stress could activate the acetylation of E2F1 at K125. Further analysis indicated that the acetylation of E2F1 at K125 could facilitate its interaction with the promoter of FAS and upregulate the levels of Fas. Furthermore, the acetylation of E2F1 attenuated its interaction with p53, thus leading to the transactivation of BAX. The upregulation of Fas and Bax activated the cleavage of caspase-3 and facilitated the apoptosis of HCC cells experiencing serum stress. Collectively, our findings indicated that the acetylation of E2F1 at K125 under serum stress leads to a functional change and a new role as an executor of cell death instead of an oncoprotein.
Collapse
Affiliation(s)
- Zejun Fang
- Central Laboratory, Sanmen People's Hospital, Sanmen 317100, China; Central Laboratory, Sanmenwan Branch, the First Affiliated Hospital, Zhejiang University School of Medicine, Sanmen 317100, China.
| | - Chaoju Gong
- Central Laboratory, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Yanyan Hu
- Department of Laboratory Medicine, Sanmen People's Hospital, Sanmen, 317100, China
| | - Tingting Cui
- Department of Ultrasound, Taizhou Traditional Chinese Medicine Hospital, Jiaojiang, 318000, China
| | - Min Lin
- Central Laboratory, Sanmen People's Hospital, Sanmen 317100, China
| | - Sha Lin
- Department of Laboratory Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Ming Ye
- Department of General Surgery, Sanmen People's Hospital, Sanmen 317100, China.
| |
Collapse
|
4
|
Krushkal J, Zhao Y, Roney K, Zhu W, Brooks A, Wilsker D, Parchment RE, McShane LM, Doroshow JH. Association of changes in expression of HDAC and SIRT genes after drug treatment with cancer cell line sensitivity to kinase inhibitors. Epigenetics 2024; 19:2309824. [PMID: 38369747 PMCID: PMC10878021 DOI: 10.1080/15592294.2024.2309824] [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: 07/24/2023] [Accepted: 01/14/2024] [Indexed: 02/20/2024] Open
Abstract
Histone deacetylases (HDACs) and sirtuins (SIRTs) are important epigenetic regulators of cancer pathways. There is a limited understanding of how transcriptional regulation of their genes is affected by chemotherapeutic agents, and how such transcriptional changes affect tumour sensitivity to drug treatment. We investigated the concerted transcriptional response of HDAC and SIRT genes to 15 approved antitumor agents in the NCI-60 cancer cell line panel. Antitumor agents with diverse mechanisms of action induced upregulation or downregulation of multiple HDAC and SIRT genes. HDAC5 was upregulated by dasatinib and erlotinib in the majority of the cell lines. Tumour cell line sensitivity to kinase inhibitors was associated with upregulation of HDAC5, HDAC1, and several SIRT genes. We confirmed changes in HDAC and SIRT expression in independent datasets. We also experimentally validated the upregulation of HDAC5 mRNA and protein expression by dasatinib in the highly sensitive IGROV1 cell line. HDAC5 was not upregulated in the UACC-257 cell line resistant to dasatinib. The effects of cancer drug treatment on expression of HDAC and SIRT genes may influence chemosensitivity and may need to be considered during chemotherapy.
Collapse
Affiliation(s)
- Julia Krushkal
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - Yingdong Zhao
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - Kyle Roney
- Department of Biostatistics and Bioinformatics, George Washington University, Washington, DC, USA
| | - Weimin Zhu
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Alan Brooks
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Deborah Wilsker
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Ralph E. Parchment
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Lisa M. McShane
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - James H. Doroshow
- Division of Cancer Treatment and Diagnosis and Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
5
|
Duan X, Xing Z, Qiao L, Qin S, Zhao X, Gong Y, Li X. The role of histone post-translational modifications in cancer and cancer immunity: functions, mechanisms and therapeutic implications. Front Immunol 2024; 15:1495221. [PMID: 39620228 PMCID: PMC11604627 DOI: 10.3389/fimmu.2024.1495221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/18/2024] [Indexed: 12/11/2024] Open
Abstract
Histones play crucial roles in both promoting and repressing gene expression, primarily regulated through post-translational modifications (PTMs) at specific amino acid residues. Histone PTMs, including methylation, acetylation, ubiquitination, phosphorylation, lactylation, butyrylation, and propionylation, act as important epigenetic markers. These modifications influence not only chromatin compaction but also gene expression. Their importance extends to the treatment and prevention of various human diseases, particularly cancer, due to their involvement in key cellular processes. Abnormal histone modifications and the enzymes responsible for these alterations often serve as critical drivers in tumor cell proliferation, invasion, apoptosis, and stemness. This review introduces key histone PTMs and the enzymes responsible for these modifications, examining their impact on tumorigenesis and cancer progression. Furthermore, it explores therapeutic strategies targeting histone PTMs and offers recommendations for identifying new potential therapeutic targets.
Collapse
Affiliation(s)
- Xiaohong Duan
- School of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, Tianjin, China
- Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, Tianjin, China
- Medical School, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Zhiyao Xing
- Tianjin University and Health-Biotech United Group Joint Laboratory of Innovative Drug Development and Translational Medicine, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
- Department of Respiratory Medicine, Jinnan Hospital, Tianjin University, Tianjin, China
- Department of Respiratory Medicine, Tianjin Jinnan Hospital, Tianjin, China
| | - Lu Qiao
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shan Qin
- Tianjin University and Health-Biotech United Group Joint Laboratory of Innovative Drug Development and Translational Medicine, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Xuejing Zhao
- Tianjin University and Health-Biotech United Group Joint Laboratory of Innovative Drug Development and Translational Medicine, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Yanhua Gong
- School of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, Tianjin, China
- Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, Tianjin, China
- Medical School, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Xueren Li
- Department of Respiratory Medicine, Jinnan Hospital, Tianjin University, Tianjin, China
- Department of Respiratory Medicine, Tianjin Jinnan Hospital, Tianjin, China
| |
Collapse
|
6
|
Lu B, Qiu R, Wei J, Wang L, Zhang Q, Li M, Zhan X, Chen J, Hsieh IY, Yang C, Zhang J, Sun Z, Zhu Y, Jiang T, Zhu H, Li J, Zhao W. Phase separation of phospho-HDAC6 drives aberrant chromatin architecture in triple-negative breast cancer. NATURE CANCER 2024; 5:1622-1640. [PMID: 39198689 DOI: 10.1038/s43018-024-00816-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 07/30/2024] [Indexed: 09/01/2024]
Abstract
How dysregulated liquid-liquid phase separation (LLPS) contributes to the oncogenesis of female triple-negative breast cancer (TNBC) remains unknown. Here we demonstrate that phosphorylated histone deacetylase 6 (phospho-HDAC6) forms LLPS condensates in the nuclei of TNBC cells that are essential for establishing aberrant chromatin architecture. The disordered N-terminal domain and phosphorylated residue of HDAC6 facilitate effective LLPS, whereas nuclear export regions exert a negative dominant effect. Through phase-separation-based screening, we identified Nexturastat A as a specific disruptor of phospho-HDAC6 condensates, which effectively suppresses tumor growth. Mechanistically, importin-β interacts with phospho-HDAC6, promoting its translocation to the nucleus, where 14-3-3θ mediates the condensate formation. Disruption of phospho-HDAC6 LLPS re-established chromatin compartments and topologically associating domain boundaries, leading to disturbed chromatin loops. The phospho-HDAC6-induced aberrant chromatin architecture affects chromatin accessibility, histone acetylation, RNA polymerase II elongation and transcriptional profiles in TNBC. This study demonstrates phospho-HDAC6 LLPS as an emerging mechanism underlying the dysregulation of chromatin architecture in TNBC.
Collapse
Affiliation(s)
- Bing Lu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ru Qiu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Jiatian Wei
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Li Wang
- Department of Anesthesiology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Qinkai Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Mingsen Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Xiudan Zhan
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Jian Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - I-Yun Hsieh
- Shunde Hospital (The First People's Hospital of Shunde), Southern Medical University, Foshan, China
| | - Ciqiu Yang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jing Zhang
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Zicheng Sun
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Yifan Zhu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Tao Jiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Han Zhu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Jie Li
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, China.
- Department of Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Wei Zhao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
7
|
Chakraborty S, Nandi P, Mishra J, Niharika, Roy A, Manna S, Baral T, Mishra P, Mishra PK, Patra SK. Molecular mechanisms in regulation of autophagy and apoptosis in view of epigenetic regulation of genes and involvement of liquid-liquid phase separation. Cancer Lett 2024; 587:216779. [PMID: 38458592 DOI: 10.1016/j.canlet.2024.216779] [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: 01/13/2024] [Revised: 02/19/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024]
Abstract
Cellular physiology is critically regulated by multiple signaling nexuses, among which cell death mechanisms play crucial roles in controlling the homeostatic landscape at the tissue level within an organism. Apoptosis, also known as programmed cell death, can be induced by external and internal stimuli directing the cells to commit suicide in unfavourable conditions. In contrast, stress conditions like nutrient deprivation, infection and hypoxia trigger autophagy, which is lysosome-mediated processing of damaged cellular organelle for recycling of the degraded products, including amino acids. Apparently, apoptosis and autophagy both are catabolic and tumor-suppressive pathways; apoptosis is essential during development and cancer cell death, while autophagy promotes cell survival under stress. Moreover, autophagy plays dual role during cancer development and progression by facilitating the survival of cancer cells under stressed conditions and inducing death in extreme adversity. Despite having two different molecular mechanisms, both apoptosis and autophagy are interconnected by several crosslinking intermediates. Epigenetic modifications, such as DNA methylation, post-translational modification of histone tails, and miRNA play a pivotal role in regulating genes involved in both autophagy and apoptosis. Both autophagic and apoptotic genes can undergo various epigenetic modifications and promote or inhibit these processes under normal and cancerous conditions. Epigenetic modifiers are uniquely important in controlling the signaling pathways regulating autophagy and apoptosis. Therefore, these epigenetic modifiers of both autophagic and apoptotic genes can act as novel therapeutic targets against cancers. Additionally, liquid-liquid phase separation (LLPS) also modulates the aggregation of misfolded proteins and provokes autophagy in the cytosolic environment. This review deals with the molecular mechanisms of both autophagy and apoptosis including crosstalk between them; emphasizing epigenetic regulation, involvement of LLPS therein, and possible therapeutic approaches against cancers.
Collapse
Affiliation(s)
- Subhajit Chakraborty
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Piyasa Nandi
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Jagdish Mishra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Niharika
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Soumen Manna
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Tirthankar Baral
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Prahallad Mishra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bypass Road, Bhauri, Bhopal, 462 030, MP, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India.
| |
Collapse
|
8
|
Yu L, Cao H, Yang JW, Meng WX, Yang C, Wang JT, Yu MM, Wang BS. HDAC5-mediated PRAME regulates the proliferation, migration, invasion, and EMT of laryngeal squamous cell carcinoma via the PI3K/AKT/mTOR signaling pathway. Open Med (Wars) 2023; 18:20230665. [PMID: 36910848 PMCID: PMC9999116 DOI: 10.1515/med-2023-0665] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/29/2022] [Accepted: 01/30/2023] [Indexed: 03/11/2023] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is an aggressive and lethal malignant neoplasm with extremely poor prognoses. Accumulating evidence has indicated that preferentially expressed antigen in melanoma (PRAME) is correlated with several kinds of cancers. However, there is little direct evidence to substantiate the biological function of PRAME in LSCC. The purpose of the current study is to explore the oncogenic role of PRAME in LSCC. PRAME expression was analyzed in 57 pairs of LSCC tumor tissue samples through quantitative real-time PCR, and the correlation between PRAME and clinicopathological features was analyzed. The result indicated that PRAME was overexpressed in the LSCC patients and correlated with the TNM staging and lymphatic metastasis. The biological functions and molecular mechanism of PRAME in LSCC progression were investigated through in vitro and in vivo assays. Functional studies confirmed that PRAME facilitated the proliferation, invasion, migration, and epithelial-mesenchymal transition of LSCC cells, and PRAME also promoted tumor growth in vivo. HDAC5 was identified as an upstream regulator that can affect the expression of PRAME. Moreover, PRAME played the role at least partially by activating PI3K/AKT/mTOR pathways. The above findings elucidate that PRAME may be a valuable oncogene target, contributing to the diagnosis and therapy of LSCC.
Collapse
Affiliation(s)
- Lei Yu
- Department of Otorhinolaryngology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huan Cao
- Department of Otorhinolaryngology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jian-Wang Yang
- Department of Otorhinolaryngology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wen-Xia Meng
- Department of Otorhinolaryngology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chuan Yang
- Department of Otorhinolaryngology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jian-Tao Wang
- Department of Otorhinolaryngology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Miao-Miao Yu
- Department of Otorhinolaryngology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Bao-Shan Wang
- Department of Otorhinolaryngology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
9
|
Ye Y, Ma Y, Kong M, Wang Z, Sun K, Li F. Effects of Dietary Phytochemicals on DNA Damage in Cancer Cells. Nutr Cancer 2023; 75:761-775. [PMID: 36562548 DOI: 10.1080/01635581.2022.2157024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
With the increasing incidence of cancer worldwide, the prevention and treatment of cancer have garnered considerable scientific attention. Traditional chemotherapeutic drugs are highly toxic and associated with substantial side effects; therefore, there is an urgent need for developing new therapeutic agents. Dietary phytochemicals are important in tumor prevention and treatment because of their low toxicity and side effects at low concentrations; however, their exact mechanisms of action remain obscure. DNA damage is mainly caused by physical or chemical factors in the environment, such as ultraviolet light, alkylating agents and reactive oxygen species that cause changes in the DNA structure of cells. Several phytochemicals have been shown inhibit the occurrence and development of tumors by inducing DNA damage. This article reviews the advances in phytochemical research; particularly regarding the mechanisms related to DNA damage and provide a theoretical basis for future chemoprophylaxis research.
Collapse
Affiliation(s)
- Yang Ye
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ying Ma
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Mei Kong
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhihua Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Kang Sun
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Fang Li
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
10
|
Gao Y, Liu Y, Zheng D, Ho C, Wen D, Sun J, Huang L, Liu Y, Li Q, Zhang Y. HDAC5-mediated Smad7 silencing through MEF2A is critical for fibroblast activation and hypertrophic scar formation. Int J Biol Sci 2022; 18:5724-5739. [PMID: 36263180 PMCID: PMC9576526 DOI: 10.7150/ijbs.76140] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/25/2022] [Indexed: 01/12/2023] Open
Abstract
Transforming growth factor-β (TGF-β) signaling plays a key role in excessive fibrosis. As a class IIa family histone deacetylase (HDAC), HDAC5 shows a close relationship with TGF-β signaling and fibrosis. However, the effect and regulatory mechanism of HDAC5 in hypertrophic scar (HS) formation remain elusive. We show that HDAC5 was overexpressed in HS tissues and depletion of HDAC5 attenuated HS formation in vivo and inhibited fibroblast activation in vitro. HDAC5 knockdown (KD) significantly downregulated TGF-β1 induced Smad2/3 phosphorylation and increased Smad7 expression. Meanwhile, Smad7 KD rescued the Smad2/3 phosphorylation downregulation and scar hyperplasia inhibition mediated by HDAC5 deficiency. Luciferase reporter assays and ChIP-qPCR assays revealed that HDAC5 interacts with myocyte enhancer factor 2A (MEF2A) suppressing MEF2A binding to the Smad7 promoter region, which results in Smad7 promoter activity repression. HDAC4/5 inhibitor, LMK235, significantly alleviated hypertrophic scar formation. Our study provides clues for the development of HDAC5 targeting strategies for the therapy or prophylaxis of fibrotic diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qingfeng Li
- ✉ Corresponding authors: Yifan Zhang and Qingfeng Li. Address: 639 Zhizaoju Road, Depart of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China. Tel: +86-21-23271699-5124; Fax: +86-21-63089567; E-mail addresses: (Yifan Zhang), (Qingfeng Li)
| | - Yifan Zhang
- ✉ Corresponding authors: Yifan Zhang and Qingfeng Li. Address: 639 Zhizaoju Road, Depart of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China. Tel: +86-21-23271699-5124; Fax: +86-21-63089567; E-mail addresses: (Yifan Zhang), (Qingfeng Li)
| |
Collapse
|
11
|
Yang B, Borgeaud AC, Buřičová M, Aeschbach L, Rodríguez-Lima O, Ruiz Buendía GA, Cinesi C, Taylor AS, Baubec T, Dion V. Expanded CAG/CTG repeats resist gene silencing mediated by targeted epigenome editing. Hum Mol Genet 2022; 31:386-398. [PMID: 34494094 PMCID: PMC8825355 DOI: 10.1093/hmg/ddab255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 05/08/2021] [Accepted: 08/25/2021] [Indexed: 11/14/2022] Open
Abstract
Expanded CAG/CTG repeat disorders affect over 1 in 2500 individuals worldwide. Potential therapeutic avenues include gene silencing and modulation of repeat instability. However, there are major mechanistic gaps in our understanding of these processes, which prevent the rational design of an efficient treatment. To address this, we developed a novel system, ParB/ANCHOR-mediated Inducible Targeting (PInT), in which any protein can be recruited at will to a GFP reporter containing an expanded CAG/CTG repeat. Previous studies have implicated the histone deacetylase HDAC5 and the DNA methyltransferase DNMT1 as modulators of repeat instability via mechanisms that are not fully understood. Using PInT, we found no evidence that HDAC5 or DNMT1 modulate repeat instability upon targeting to the expanded repeat, suggesting that their effect is independent of local chromatin structure. Unexpectedly, we found that expanded CAG/CTG repeats reduce the effectiveness of gene silencing mediated by targeting HDAC5 and DNMT1. The repeat-length effect in gene silencing by HDAC5 was abolished by a small molecule inhibitor of HDAC3. Our results have important implications on the design of epigenome editing approaches for expanded CAG/CTG repeat disorders. PInT is a versatile synthetic system to study the effect of any sequence of interest on epigenome editing.
Collapse
Affiliation(s)
- Bin Yang
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Alicia C Borgeaud
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Marcela Buřičová
- UK Dementia Research Institute at Cardiff University, Cardiff, UK
| | - Lorène Aeschbach
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Oscar Rodríguez-Lima
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Gustavo A Ruiz Buendía
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Cinzia Cinesi
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Alysha S Taylor
- UK Dementia Research Institute at Cardiff University, Cardiff, UK
| | - Tuncay Baubec
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Vincent Dion
- UK Dementia Research Institute at Cardiff University, Cardiff, UK
| |
Collapse
|
12
|
Abstract
Autophagy is an intracellular catabolic degradative process in which damaged cellular organelles, unwanted proteins and different cytoplasmic components get recycled to maintain cellular homeostasis or metabolic balance. During autophagy, a double membrane vesicle is formed to engulf these cytosolic materials and fuse to lysosomes wherein the entire cargo degrades to be used again. Because of this unique recycling ability of cells, autophagy is a universal stress response mechanism. Dysregulation of autophagy leads to several diseases, including cancer, neurodegeneration and microbial infection. Thus, autophagy machineries have become targets for therapeutics. This chapter provides an overview of the paradoxical role of autophagy in tumorigenesis in the perspective of metabolism.
Collapse
Affiliation(s)
- Sweta Sikder
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Atanu Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Homi Bhaba National Institute, Mumbai, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Homi Bhaba National Institute, Mumbai, India
| | - Tapas K Kundu
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India.
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, India.
| |
Collapse
|
13
|
Shi Y, Shen HM, Gopalakrishnan V, Gordon N. Epigenetic Regulation of Autophagy Beyond the Cytoplasm: A Review. Front Cell Dev Biol 2021; 9:675599. [PMID: 34195194 PMCID: PMC8237754 DOI: 10.3389/fcell.2021.675599] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a highly conserved catabolic process induced under various stress conditions to protect the cell from harm and allow survival in the face of nutrient- or energy-deficient states. Regulation of autophagy is complex, as cells need to adapt to a continuously changing microenvironment. It is well recognized that the AMPK and mTOR signaling pathways are the main regulators of autophagy. However, various other signaling pathways have also been described to regulate the autophagic process. A better understanding of these complex autophagy regulatory mechanisms will allow the discovery of new potential therapeutic targets. Here, we present a brief overview of autophagy and its regulatory pathways with emphasis on the epigenetic control mechanisms.
Collapse
Affiliation(s)
- Yin Shi
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, China
| | - Han-Ming Shen
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Vidya Gopalakrishnan
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nancy Gordon
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
14
|
Yang J, Gong C, Ke Q, Fang Z, Chen X, Ye M, Xu X. Insights Into the Function and Clinical Application of HDAC5 in Cancer Management. Front Oncol 2021; 11:661620. [PMID: 34178647 PMCID: PMC8222663 DOI: 10.3389/fonc.2021.661620] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/18/2021] [Indexed: 12/20/2022] Open
Abstract
Histone deacetylase 5 (HDAC5) is a class II HDAC. Aberrant expression of HDAC5 has been observed in multiple cancer types, and its functions in cell proliferation and invasion, the immune response, and maintenance of stemness have been widely studied. HDAC5 is considered as a reliable therapeutic target for anticancer drugs. In light of recent findings regarding the role of epigenetic reprogramming in tumorigenesis, in this review, we provide an overview of the expression, biological functions, regulatory mechanisms, and clinical significance of HDAC5 in cancer.
Collapse
Affiliation(s)
- Jun Yang
- Department of Orthopedic Surgery, Sanmen People's Hospital of Zhejiang Province, Sanmenwan Branch of the First Affiliated Hospital, College of Medicine, Zhejiang University, Sanmen, China
| | - Chaoju Gong
- Central Laboratory, The Municipal Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qinjian Ke
- Central Laboratory, Sanmen People's Hospital of Zhejiang Province, Sanmenwan Branch of the First Affiliated Hospital, College of Medicine, Zhejiang University, Sanmen, China
| | - Zejun Fang
- Central Laboratory, Sanmen People's Hospital of Zhejiang Province, Sanmenwan Branch of the First Affiliated Hospital, College of Medicine, Zhejiang University, Sanmen, China
| | - Xiaowen Chen
- Department of Pathophysiology, Zunyi Medical University, Zunyi, China
| | - Ming Ye
- Department of General Surgery, Sanmen People's Hospital of Zhejiang Province, Sanmenwan Branch of the First Affiliated Hospital, College of Medicine, Zhejiang University, Sanmen, China
| | - Xi Xu
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
15
|
Vodnala M, Choi EB, Fong YW. Low complexity domains, condensates, and stem cell pluripotency. World J Stem Cells 2021; 13:416-438. [PMID: 34136073 PMCID: PMC8176841 DOI: 10.4252/wjsc.v13.i5.416] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Biological reactions require self-assembly of factors in the complex cellular milieu. Recent evidence indicates that intrinsically disordered, low-complexity sequence domains (LCDs) found in regulatory factors mediate diverse cellular processes from gene expression to DNA repair to signal transduction, by enriching specific biomolecules in membraneless compartments or hubs that may undergo liquid-liquid phase separation (LLPS). In this review, we discuss how embryonic stem cells take advantage of LCD-driven interactions to promote cell-specific transcription, DNA damage response, and DNA repair. We propose that LCD-mediated interactions play key roles in stem cell maintenance and safeguarding genome integrity.
Collapse
Affiliation(s)
- Munender Vodnala
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Eun-Bee Choi
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Yick W Fong
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
- Harvard Stem Cell Institute, Cambridge, MA 02138, United States.
| |
Collapse
|
16
|
Bhol CS, Panigrahi DP, Praharaj PP, Mahapatra KK, Patra S, Mishra SR, Behera BP, Bhutia SK. Epigenetic modifications of autophagy in cancer and cancer therapeutics. Semin Cancer Biol 2020; 66:22-33. [DOI: 10.1016/j.semcancer.2019.05.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/09/2019] [Accepted: 05/30/2019] [Indexed: 12/30/2022]
|
17
|
Chidamide, a histone deacetylase inhibitor, inhibits autophagy and exhibits therapeutic implication in chronic lymphocytic leukemia. Aging (Albany NY) 2020; 12:16083-16098. [PMID: 32855355 PMCID: PMC7485718 DOI: 10.18632/aging.103536] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 06/04/2020] [Indexed: 01/28/2023]
Abstract
Novel agents have made the management of chronic lymphocytic leukemia (CLL) more promising and personalized. However, long-term treatment is still warranted which may result in toxicity and resistance. Thus, new combination therapy may help achieve deeper remission and limited-duration therapy. Histone deacetylase inhibitors (HDACi) can affect many tumors by modulating key biological functions including autophagy. Studies have shown that some novel targeted agents including ibrutinib induce autophagy. This study aimed to explore the effect of oral HDAC inhibitor, chidamide, on CLL cells as well as the role of autophagy in this process. Here, we showed that autophagy flux in CLL cells was inhibited by chidamide via post-transcriptional modulation and chidamide had cytostatic and cytotoxic effects on CLL cells. Besides, the pro-survival role of autophagy in CLL cells was validated by using autophagy inhibitor and knocking down critical autophagy gene. Notably, a combination of chidamide and ibrutinib showed significant synergism and downregulated ibrutinib-induced autophagy. This work highlights the therapeutic potential of chidamide via its effect on autophagy, especially in combination with ibrutinib.
Collapse
|
18
|
Tan H, Zhang S, Zhang J, Zhu L, Chen Y, Yang H, Chen Y, An Y, Liu B. Long non-coding RNAs in gastric cancer: New emerging biological functions and therapeutic implications. Am J Cancer Res 2020; 10:8880-8902. [PMID: 32754285 PMCID: PMC7392009 DOI: 10.7150/thno.47548] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/28/2020] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) is currently the fourth most common malignancy and the third leading cause of cancer-related deaths worldwide. Long non-coding RNAs (lncRNAs), transcriptional products with more than 200 nucleotides, are not as well-characterized as protein-coding RNAs. Accumulating evidence has recently revealed that maladjustments of diverse lncRNAs may play key roles in multiple genetic and epigenetic phenomena in GC, affecting all aspects of cellular homeostasis, such as proliferation, migration, and stemness. However, the full extent of their functionality remains to be clarified. Considering the lack of viable biomarkers and therapeutic targets, future research should be focused on unravelling the intricate relationships between lncRNAs and GC that can be translated from bench to clinic. Here, we summarized the state-of-the-art advances in lncRNAs and their biological functions in GC, and we further discuss their potential diagnostic and therapeutic roles. We aim to shed light on the interrelationships between lncRNAs and GC with respect to their potential therapeutic applications. With better understanding of these relationships, the biological functions of lncRNAs in GC development will be exploitable, and promising new strategies developed for the prevention and treatment of GC.
Collapse
|
19
|
Pant K, Peixoto E, Richard S, Gradilone SA. Role of Histone Deacetylases in Carcinogenesis: Potential Role in Cholangiocarcinoma. Cells 2020; 9:cells9030780. [PMID: 32210140 PMCID: PMC7140894 DOI: 10.3390/cells9030780] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/05/2020] [Accepted: 03/17/2020] [Indexed: 12/19/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a highly invasive and metastatic form of carcinoma with bleak prognosis due to limited therapies, frequent relapse, and chemotherapy resistance. There is an urgent need to identify the molecular regulators of CCA in order to develop novel therapeutics and advance diseases diagnosis. Many cellular proteins including histones may undergo a series of enzyme-mediated post-translational modifications including acetylation, methylation, phosphorylation, sumoylation, and crotonylation. Histone deacetylases (HDACs) play an important role in regulating epigenetic maintenance and modifications of their targets, which in turn exert critical impacts on chromatin structure, gene expression, and stability of proteins. As such, HDACs constitute a group of potential therapeutic targets for CCA. The aim of this review was to summarize the role that HDACs perform in regulating epigenetic changes, tumor development, and their potential as therapeutic targets for CCA.
Collapse
Affiliation(s)
- Kishor Pant
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; (K.P.); (E.P.); (S.R.)
| | - Estanislao Peixoto
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; (K.P.); (E.P.); (S.R.)
| | - Seth Richard
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; (K.P.); (E.P.); (S.R.)
| | - Sergio A. Gradilone
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; (K.P.); (E.P.); (S.R.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Correspondence:
| |
Collapse
|
20
|
dos Santos PWDS, Machado ART, De Grandis RA, Ribeiro DL, Tuttis K, Morselli M, Aissa AF, Pellegrini M, Antunes LMG. Transcriptome and DNA methylation changes modulated by sulforaphane induce cell cycle arrest, apoptosis, DNA damage, and suppression of proliferation in human liver cancer cells. Food Chem Toxicol 2020; 136:111047. [DOI: 10.1016/j.fct.2019.111047] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/30/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
|
21
|
Epigenetic Control of Autophagy in Cancer Cells: A Key Process for Cancer-Related Phenotypes. Cells 2019; 8:cells8121656. [PMID: 31861179 PMCID: PMC6952790 DOI: 10.3390/cells8121656] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/19/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
Although autophagy is a well-known and extensively described cell pathway, numerous studies have been recently interested in studying the importance of its regulation at different molecular levels, including the translational and post-translational levels. Therefore, this review focuses on the links between autophagy and epigenetics in cancer and summarizes the. following: (i) how ATG genes are regulated by epigenetics, including DNA methylation and post-translational histone modifications; (ii) how epidrugs are able to modulate autophagy in cancer and to alter cancer-related phenotypes (proliferation, migration, invasion, tumorigenesis, etc.) and; (iii) how epigenetic enzymes can also regulate autophagy at the protein level. One noteable observation was that researchers most often reported conclusions about the regulation of the autophagy flux, following the use of epidrugs, based only on the analysis of LC3B-II form in treated cells. However, it is now widely accepted that an increase in LC3B-II form could be the consequence of an induction of the autophagy flux, as well as a block in the autophagosome-lysosome fusion. Therefore, in our review, all the published results describing a link between epidrugs and autophagy were systematically reanalyzed to determine whether autophagy flux was indeed increased, or inhibited, following the use of these potentially new interesting treatments targeting the autophagy process. Altogether, these recent data strongly support the idea that the determination of autophagy status could be crucial for future anticancer therapies. Indeed, the use of a combination of epidrugs and autophagy inhibitors could be beneficial for some cancer patients, whereas, in other cases, an increase of autophagy, which is frequently observed following the use of epidrugs, could lead to increased autophagy cell death.
Collapse
|
22
|
Myoferlin Contributes to the Metastatic Phenotype of Pancreatic Cancer Cells by Enhancing Their Migratory Capacity through the Control of Oxidative Phosphorylation. Cancers (Basel) 2019; 11:cancers11060853. [PMID: 31248212 PMCID: PMC6628295 DOI: 10.3390/cancers11060853] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/13/2019] [Accepted: 06/16/2019] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies with an overall survival of 5% and is the second cause of death by cancer, mainly linked to its high metastatic aggressiveness. Accordingly, understanding the mechanisms sustaining the PDAC metastatic phenotype remains a priority. In this study, we generated and used a murine in vivo model to select clones from the human Panc-1 PDAC cell line that exhibit a high propensity to seed and metastasize into the liver. We showed that myoferlin, a protein previously reported to be overexpressed in PDAC, is significantly involved in the migratory abilities of the selected cells. We first report that highly metastatic Panc-1 clones expressed a significantly higher myoferlin level than the corresponding low metastatic ones. Using scratch wound and Boyden’s chamber assays, we show that cells expressing a high myoferlin level have higher migratory potential than cells characterized by a low myoferlin abundance. Moreover, we demonstrate that myoferlin silencing leads to a migration decrease associated with a reduction of mitochondrial respiration. Since mitochondrial oxidative phosphorylation has been shown to be implicated in the tumor progression and dissemination, our data identify myoferlin as a valid potential therapeutic target in PDAC.
Collapse
|
23
|
Costanza B, Rademaker G, Tiamiou A, De Tullio P, Leenders J, Blomme A, Bellier J, Bianchi E, Turtoi A, Delvenne P, Bellahcène A, Peulen O, Castronovo V. Transforming growth factor beta-induced, an extracellular matrix interacting protein, enhances glycolysis and promotes pancreatic cancer cell migration. Int J Cancer 2019; 145:1570-1584. [PMID: 30834519 DOI: 10.1002/ijc.32247] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 02/02/2019] [Accepted: 02/25/2019] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a deadly malignancy with no efficient therapy available up-to-date. Glycolysis is the main provider of energetic substrates to sustain cancer dissemination of PDAC. Accordingly, altering the glycolytic pathway is foreseen as a sound approach to trigger pancreatic cancer regression. Here, we show for the first time that high transforming growth factor beta-induced (TGFBI) expression in PDAC patients is associated with a poor outcome. We demonstrate that, although usually secreted by stromal cells, PDAC cells synthesize and secrete TGFBI in quantity correlated with their migratory capacity. Mechanistically, we show that TGFBI activates focal adhesion kinase signaling pathway through its binding to integrin αVβ5, leading to a significant enhancement of glycolysis and to the acquisition of an invasive phenotype. Finally, we show that TGFBI silencing significantly inhibits PDAC tumor development in a chick chorioallantoic membrane assay model. Our study highlights TGFBI as an oncogenic extracellular matrix interacting protein that bears the potential to serve as a target for new anti-PDAC therapeutic strategies.
Collapse
Affiliation(s)
- Brunella Costanza
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Gilles Rademaker
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Assia Tiamiou
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Pascal De Tullio
- Center for Interdisciplinary Research on Medicines, Metabolomics Group, University of Liège, Liège, Belgium
| | - Justine Leenders
- Center for Interdisciplinary Research on Medicines, Metabolomics Group, University of Liège, Liège, Belgium
| | - Arnaud Blomme
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Justine Bellier
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Elettra Bianchi
- Department of Pathology, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Andrei Turtoi
- Tumor Microenvironment and Resistance to Treatment Laboratory, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
| | - Philippe Delvenne
- Department of Pathology, University Hospital (CHU), University of Liège, Liège, Belgium.,Laboratory of Experimental Pathology, GIGA Cancer, University of Liège, Liège, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| |
Collapse
|
24
|
Koosha S, Mohamed Z, Sinniah A, Alshawsh MA. Investigation into the Molecular Mechanisms underlying the Anti-proliferative and Anti-tumorigenesis activities of Diosmetin against HCT-116 Human Colorectal Cancer. Sci Rep 2019; 9:5148. [PMID: 30914796 PMCID: PMC6435658 DOI: 10.1038/s41598-019-41685-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 03/08/2019] [Indexed: 01/06/2023] Open
Abstract
Diosmetin (Dis) is a bioflavonoid with cytotoxicity properties against variety of cancer cells including hepatocarcinoma, breast and colorectal (CRC) cancer. The exact mechanism by which Dis acts against CRC however, still remains unclear, hence in this study, we investigated the possible molecular mechanisms of Dis in CRC cell line, HCT-116. Here, we monitored the viability of HCT-116 cells in the presence of Dis and investigated the underlying mechanism of Dis against HCT-116 cells at the gene and protein levels using NanoString and proteome profiler array technologies. Findings demonstrated that Dis exhibits greater cytotoxic effects towards HCT-116 CRC cells (IC50 = 3.58 ± 0.58 µg/ml) as compared to the normal colon CCD-841 cells (IC50 = 51.95 ± 0.11 µg/ml). Arrests of the cells in G2/M phase confirms the occurrence of mitotic disruption via Dis. Activation of apoptosis factors such as Fas and Bax at the gene and protein levels along with the release of Cytochrome C from mitochondria and cleavage of Caspase cascades indicate the presence of turbulence as a result of apoptosis induction in Dis-treated cells. Moreover, NF-ƙB translocation was inhibited in Dis-treated cells. Our results indicate that Dis can target HCT-116 cells through the mitotic disruption and apoptosis induction.
Collapse
Affiliation(s)
- Sanaz Koosha
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Zahurin Mohamed
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ajantha Sinniah
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Mohammed A Alshawsh
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
25
|
Human colon cancer cells highly express myoferlin to maintain a fit mitochondrial network and escape p53-driven apoptosis. Oncogenesis 2019; 8:21. [PMID: 30850580 PMCID: PMC6408501 DOI: 10.1038/s41389-019-0130-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 02/18/2019] [Indexed: 12/24/2022] Open
Abstract
Colon adenocarcinoma is the third most commonly diagnosed cancer and the second deadliest one. Metabolic reprogramming, described as an emerging hallmark of malignant cells, includes the predominant use of glycolysis to produce energy. Recent studies demonstrated that mitochondrial electron transport chain inhibitor reduced colon cancer tumour growth. Accumulating evidence show that myoferlin, a member of the ferlin family, is highly expressed in several cancer types, where it acts as a tumour promoter and participates in the metabolic rewiring towards oxidative metabolism. In this study, we showed that myoferlin expression in colon cancer lesions is associated with low patient survival and is higher than in non-tumoural adjacent tissue. Human colon cancer cells silenced for myoferlin exhibit a reduced oxidative phosphorylation activity associated with mitochondrial fission leading, ROS accumulation, decreased cell growth, and increased apoptosis. We observed the triggering of a DNA damage response culminating to a cell cycle arrest in wild-type p53 cells. The use of a p53 null cell line or a compound able to restore p53 activity (Prima-1) reverted the effects induced by myoferlin silencing, confirming the involvement of p53. The recent identification of a compound interacting with a myoferlin C2 domain and bearing anticancer potency identifies, together with our demonstration, this protein as a suitable new therapeutic target in colon cancer.
Collapse
|
26
|
Wanek J, Gaisberger M, Beyreis M, Mayr C, Helm K, Primavesi F, Jäger T, Di Fazio P, Jakab M, Wagner A, Neureiter D, Kiesslich T. Pharmacological Inhibition of Class IIA HDACs by LMK-235 in Pancreatic Neuroendocrine Tumor Cells. Int J Mol Sci 2018; 19:3128. [PMID: 30321986 PMCID: PMC6213165 DOI: 10.3390/ijms19103128] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/18/2018] [Accepted: 09/29/2018] [Indexed: 02/07/2023] Open
Abstract
Histone deacetylases (HDACs) play a key role in epigenetic mechanisms in health and disease and their dysfunction is implied in several cancer entities. Analysis of expression patterns in pancreatic neuroendocrine tumors (pNETs) indicated HDAC5 to be a potential target for future therapies. As a first step towards a possible treatment, the aim of this study was to evaluate the in vitro cellular and molecular effects of HDAC5 inhibition in pNET cells. Two pNET cell lines, BON-1 and QGP-1, were incubated with different concentrations of the selective class IIA HDAC inhibitor, LMK-235. Effects on cell viability were determined using the resazurin-assay, the caspase-assay, and Annexin-V staining. Western Blot and immunofluorescence microscopy were performed to assess the effects on HDAC5 functionality. LMK-235 lowered overall cell viability by inducing apoptosis in a dose- and time-dependent manner. Furthermore, acetylation of histone-H3 increased with higher LMK-235 concentrations, indicating functional inhibition of HDAC4/5. Immunocytochemical analysis showed that proliferative activity (phosphohistone H3 and Ki-67) decreased at highest concentrations of LMK-235 while chromogranin and somatostatin receptor 2 (SSTR2) expression increased in a dose-dependent manner. HDAC5 expression was found to be largely unaffected by LMK-235. These findings indicate LMK-235 to be a potential therapeutic approach for the development of an effective and selective pNET treatment.
Collapse
Affiliation(s)
- Julia Wanek
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
- Biomedical Sciences, Salzburg University of Applied Sciences, 5412 Puch/Salzburg, Austria.
- Department of Biosciences, Research Division of Regeneration, Stem Cell Biology and Gerontology, University of Salzburg, 5020 Salzburg, Austria.
| | - Martin Gaisberger
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria.
- Department for Radon Therapy Research, Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria.
- Gastein Research Institute, Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria.
| | - Marlena Beyreis
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria.
| | - Christian Mayr
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria.
| | - Katharina Helm
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
- Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Florian Primavesi
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria.
- Department of Surgery, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
| | - Tarkan Jäger
- Department of Surgery, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
| | - Pietro Di Fazio
- Department of Visceral Thoracic and Vascular Surgery, Philipps University Marburg, 35033 Marburg, Germany.
| | - Martin Jakab
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria.
| | - Andrej Wagner
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
- Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Tobias Kiesslich
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria.
| |
Collapse
|
27
|
Zhang E, He X, Zhang C, Su J, Lu X, Si X, Chen J, Yin D, Han L, De W. A novel long noncoding RNA HOXC-AS3 mediates tumorigenesis of gastric cancer by binding to YBX1. Genome Biol 2018; 19:154. [PMID: 30286788 PMCID: PMC6172843 DOI: 10.1186/s13059-018-1523-0] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 09/03/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Recently, increasing evidence shows that long noncoding RNAs (lncRNAs) play a significant role in human tumorigenesis. However, the function of lncRNAs in human gastric cancer remains largely unknown. RESULTS By using publicly available expression profiling data from gastric cancer and integrating bioinformatics analyses, we screen and identify a novel lncRNA, HOXC-AS3. HOXC-AS3 is significantly increased in gastric cancer tissues and is correlated with clinical outcomes of gastric cancer. In addition, HOXC-AS3 regulates cell proliferation and migration both in vitro and in vivo. RNA-seq analysis reveals that HOXC-AS3 knockdown preferentially affects genes that are linked to proliferation and migration. Mechanistically, we find that HOXC-AS3 is obviously activated by gain of H3K4me3 and H3K27ac, both in cells and in tissues. RNA pull-down mass spectrometry analysis identifies that YBX1 interacts with HOXC-AS3, and RNA-seq analysis finds a marked overlap in genes differentially expressed after YBX1 knockdown and those transcriptionally regulated by HOXC-AS3, suggesting that YBX1 participates in HOXC-AS3-mediated gene transcriptional regulation in the tumorigenesis of gastric cancer. CONCLUSIONS Together, our data demonstrate that abnormal histone modification-activated HOXC-AS3 may play important roles in gastric cancer oncogenesis and may serve as a target for gastric cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Erbao Zhang
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
| | - Xuezhi He
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Chongguo Zhang
- Department of Oncology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Jun Su
- Department of Oncology, The Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin, Jiangsu, People's Republic of China
| | - Xiyi Lu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Xinxin Si
- Huaihai Institute of Technology, Lianyungang, Jiangsu, People's Republic of China
| | - Jinfei Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Dandan Yin
- Cancer Research and Biotherapy Center, Nanjing Second Hospital, the Second Affiliated Hospital of Southeast University, Nanjing, Jiangsu, People's Republic of China.
| | - Liang Han
- Department of Oncology, Xuzhou Central Hospital, Affiliated Xuzhou Hospital, College of Medicine, Southeast University, Xuzhou, Jiangsu, People's Republic of China.
- Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.
| | - Wei De
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
28
|
Abstract
Pancreatic cancer is an aggressive and intractable malignancy with high mortality. This is due in part to a high resistance to chemotherapeutics and radiation treatment conferred by diverse regulatory mechanisms. Among these, constituents of the nuclear envelope play a significant role in regulating oncogenesis and pancreatic tumor biology, and this review focuses on three specific components and their roles in cancer. The LINC complex is a nuclear envelope component formed by proteins with SUN and KASH domains that interact in the periplasmic space of the nuclear envelope. These interactions functionally and structurally couple the cytoskeleton to chromatin and facilitates gene regulation informed by cytoplasmic activity. Furthermore, cancer cell invasiveness is impacted by LINC complex biology. The nuclear lamina is adjacent to the inner nuclear membrane of the nuclear envelope and can actively regulate chromatin in addition to providing structural integrity to the nucleus. A disrupted lamina can impart biophysical compromise to nuclear structure and function, as well as form dysfunctional micronuclei that may lead to genomic instability and chromothripsis. In close relationship to the nuclear lamina is the nuclear pore complex, a large megadalton structure that spans both outer and inner membranes of the nuclear envelope. The nuclear pore complex mediates bidirectional nucleocytoplasmic transport and is comprised of specialized proteins called nucleoporins that are overexpressed in many cancers and are diagnostic markers for oncogenesis. Furthermore, recent demonstration of gene regulatory functions for discrete nucleoporins independent of their nuclear trafficking function suggests that these proteins may contribute more to malignant phenotypes beyond serving as biomarkers. The nuclear envelope is thus a complex, intricate regulator of cell signaling, with roles in pancreatic tumorigenesis and general oncogenic transformation.
Collapse
Affiliation(s)
| | - Randolph S. Faustino
- Genetics and Genomics, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
29
|
Up-regulation of HO-1 promotes resistance of B-cell acute lymphocytic leukemia cells to HDAC4/5 inhibitor LMK-235 via the Smad7 pathway. Life Sci 2018; 207:386-394. [PMID: 29886060 DOI: 10.1016/j.lfs.2018.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/22/2018] [Accepted: 06/04/2018] [Indexed: 02/03/2023]
Abstract
PURPOSE HDAC4/5 and Smad7 are potential therapeutic targets for the onset and progression of B-cell acute lymphocytic leukemia (B-ALL) and indices for clinical prognosis. In contrast, HO-1 (heat shock protein 32) plays a key role in protecting tumor cells from apoptosis. METHODS HDAC4/5, HO-1 and Smad7 expressions in 34 newly diagnosed B-ALL cases were detected by real-time PCR and Western blot. Lentivirus and small interference RNA were used to transfect B-ALL cells. The expression of Smad7 was detected after treatment with LMK-235 or Hemin and ZnPP. Apoptosis and proliferation were evaluated by flow cytometry, CCK-8 assay and Western blot. RESULTS HDAC4/5 was overexpressed in B-ALL patients with high HO-1 levels. Increasing the concentration of HDAC4/5 inhibitor LMK-235 induced the decrease of Smad7 and HO-1 expressions and the apoptosis of B-ALL cells by suppressing the phosphorylation of AKT (Protein kinase B). Up-regulating HO-1 alleviated the decrease of Smad7 expression and enhanced B-ALL resistance to LMK-235 by activating p-AKT which reduced the apoptosis of B-ALL cells and influenced the survival of leukemia patients. Silencing Smad7 also augmented the apoptosis rate of B-ALL cells by suppressing p-AKT. CONCLUSION HO-1 played a key role in protecting tumor cells from apoptosis, and HDAC4/5 were related with the apoptosis of B-ALL cells. LMK-235 may be able to improve the poor survival of leukemia patients.
Collapse
|
30
|
Myoferlin controls mitochondrial structure and activity in pancreatic ductal adenocarcinoma, and affects tumor aggressiveness. Oncogene 2018; 37:4398-4412. [PMID: 29720728 PMCID: PMC6085282 DOI: 10.1038/s41388-018-0287-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/08/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related death. Therapeutic options remain very limited and are based on classical chemotherapies. Energy metabolism reprogramming appears as an emerging hallmark of cancer and is considered a therapeutic target with considerable potential. Myoferlin, a ferlin family member protein overexpressed in PDAC, is involved in plasma membrane biology and has a tumor-promoting function. In the continuity of our previous studies, we investigated the role of myoferlin in the context of energy metabolism in PDAC. We used selected PDAC tumor samples and PDAC cell lines together with small interfering RNA technology to study the role of myoferlin in energetic metabolism. In PDAC patients, we showed that myoferlin expression is negatively correlated with overall survival and with glycolytic activity evaluated by 18F-deoxyglucose positron emission tomography. We found out that myoferlin is more abundant in lipogenic pancreatic cancer cell lines and is required to maintain a branched mitochondrial structure and a high oxidative phosphorylation activity. The observed mitochondrial fission induced by myoferlin depletion led to a decrease of cell proliferation, ATP production, and autophagy induction, thus indicating an essential role of myoferlin for PDAC cell fitness. The metabolic phenotype switch generated by myoferlin silencing could open up a new perspective in the development of therapeutic strategies, especially in the context of energy metabolism.
Collapse
|
31
|
Li A, Liu Z, Li M, Zhou S, Xu Y, Xiao Y, Yang W. HDAC5, a potential therapeutic target and prognostic biomarker, promotes proliferation, invasion and migration in human breast cancer. Oncotarget 2018; 7:37966-37978. [PMID: 27177225 PMCID: PMC5122364 DOI: 10.18632/oncotarget.9274] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 04/26/2016] [Indexed: 12/22/2022] Open
Abstract
Purpose Histone deacetylase 5 (HDAC5) is an important protein in neural and cardiac diseases and a potential drug target. However, little is known regarding the specific role of HDAC5 in breast cancer (BC). We aimed to evaluate HDAC5 expression in human breast tumors and to determine the effects of the inhibition of HDAC5 expression in BC cells. Experimental design HDAC5 expression was evaluated in BC patients and was correlated with clinical features and with patient prognosis. Functional experiments were performed using shRNA and the selective HDAC inhibitor LMK-235 for HDAC5 knockdown and inhibition in BC cells. The synergistic effects of LMK-235 with the proteasome inhibitor bortezomib were also examined. Results HDAC5 was extensively expressed in human BC tissues, and high HDAC5 expression was associated with an inferior prognosis. Knockdown of HDAC5 inhibited cell proliferation, migration, invasion, and enhanced apoptosis. The HDAC5 inhibitor LMK-235 inhibited cell growth and induced apoptosis, while the inclusion of bortezomib synergistically enhanced the efficacy of LMK-235. Conclusions Our findings indicate that HDAC5 is a promising prognostic marker and drug target for BC and that the combination of LMK-235 and bortezomib presents a novel therapeutic strategy for BC.
Collapse
Affiliation(s)
- Anqi Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Zebing Liu
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P.R. China
| | - Ming Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Shuling Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yan Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yaoxing Xiao
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| |
Collapse
|
32
|
Xu Q, Liu X, Zhu S, Hu X, Niu H, Zhang X, Zhu D, Nesa EU, Tian K, Yuan H. Hyper-acetylation contributes to the sensitivity of chemo-resistant prostate cancer cells to histone deacetylase inhibitor Trichostatin A. J Cell Mol Med 2018; 22:1909-1922. [PMID: 29327812 PMCID: PMC5824406 DOI: 10.1111/jcmm.13475] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/30/2017] [Indexed: 11/30/2022] Open
Abstract
Therapeutic agents are urgently needed for treating metastatic castration‐refractory prostate cancer (mCRPC) that is unresponsive to androgen deprivation and chemotherapy. Our screening assays demonstrated that chemotherapy‐resistant prostate cancer (PCa) cells are more sensitive to HDAC inhibitors than paired sensitive PCa cells, as demonstrated by cell proliferation and apoptosis in vitro and in vivo. Kinetic study revealed that TSA‐induced apoptosis was significantly dependent on enhanced transcription and protein synthesis in an early stage, which subsequently caused ER stress and apoptosis. ChIP analysis indicated that TSA increased H4K16 acetylation, promoting ER stress gene transcription. The changes in Ac‐H4K16, ATF3 and ATF4 were also validated in TSA‐treated animals. Further study revealed the higher enzyme activity of HDACs and an increase in acetylated proteins in resistant cells. The higher nucleocytoplasmic acetyl‐CoA in resistant cells was responsible for elevated acetylation status of protein and a more vigorous growth state. These results strongly support the pre‐clinical application of HDAC inhibitors for treating chemotherapy‐resistant mCRPC.
Collapse
Affiliation(s)
- Qingqing Xu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Xiaofei Liu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Shiqin Zhu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Xuelei Hu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Huanmin Niu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Xiulei Zhang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Deyu Zhu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Effat Un Nesa
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Keli Tian
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Huiqing Yuan
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| |
Collapse
|
33
|
Klieser E, Urbas R, Stättner S, Primavesi F, Jäger T, Dinnewitzer A, Mayr C, Kiesslich T, Holzmann K, Di Fazio P, Neureiter D, Swierczynski S. Comprehensive immunohistochemical analysis of histone deacetylases in pancreatic neuroendocrine tumors: HDAC5 as a predictor of poor clinical outcome. Hum Pathol 2017; 65:41-52. [PMID: 28235630 DOI: 10.1016/j.humpath.2017.02.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 01/18/2017] [Accepted: 02/10/2017] [Indexed: 02/07/2023]
Abstract
Epigenetic factors contribute to carcinogenesis, tumor promotion, and chemoresistance. Histone deacetylases (HDACs) are epigenetic regulators that primarily cause chromatin compaction, leading to inaccessibility of promoter regions and eventually gene silencing. Many cancer entities feature overexpression of HDACs. Currently, the role of HDACs in pancreatic neuroendocrine tumors (pNETs) is unclear. We analyzed the expression patterns of all HDAC classes (classes I, IIA, IIB, III, and IV) in 5 human tissue microarrays representing 57 pNETs resected between 1997 and 2013 and corresponding control tissue. All pNET cases were characterized clinically and pathologically according to recent staging guidelines. The investigated cases included 32 (56.1%) female and 25 (43.9%) male pNET patients (total n=57, 47.4% immunohistochemically endocrine positive). Immunohistochemical profiling revealed a significant up-regulation of all HDAC classes in pNET versus control, with different levels of intensity and extensity ranging from 1.5- to >7-fold up-regulation. In addition, expression of several HDACs (HDAC1, HDAC2, HDAC5, HDAC11, and Sirt1) was significantly increased in G3 tumors. Correlation analysis showed a significant association between the protein expression of HDAC classes I, III, and IV and rate of the pHH3/Ki-67-associated mitotic and proliferation index. Furthermore, especially HDAC5 proved as a negative predictor of disease-free and overall survival in pNET patients. Overall, we demonstrate that specific members of all 4 HDAC classes are heterogeneously expressed in pNET. Moreover, expression of HDACs was associated with tumor grading, proliferation markers, and patient survival, therefore representing interesting new targets in pNET treatment.
Collapse
Affiliation(s)
- Eckhard Klieser
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
| | - Romana Urbas
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
| | - Stefan Stättner
- Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, 6020 Innsbruck, Austria.
| | - Florian Primavesi
- Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, 6020 Innsbruck, Austria.
| | - Tarkan Jäger
- Department of Surgery, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
| | - Adam Dinnewitzer
- Department of Surgery, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
| | - Christian Mayr
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria; Laboratory for Tumour Biology and Experimental Therapies, Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria.
| | - Tobias Kiesslich
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria; Laboratory for Tumour Biology and Experimental Therapies, Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria.
| | - Klaus Holzmann
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna 1090, Austria.
| | - Pietro Di Fazio
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University of Marburg, 35043 Marburg, Germany.
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
| | - Stefan Swierczynski
- Department of Surgery, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria.
| |
Collapse
|
34
|
Hendrick E, Peixoto P, Blomme A, Polese C, Matheus N, Cimino J, Frère A, Mouithys-Mickalad A, Serteyn D, Bettendorff L, Elmoualij B, De Tullio P, Eppe G, Dequiedt F, Castronovo V, Mottet D. Metabolic inhibitors accentuate the anti-tumoral effect of HDAC5 inhibition. Oncogene 2017; 36:4859-4874. [PMID: 28414307 DOI: 10.1038/onc.2017.103] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 12/17/2022]
Abstract
The US FDA approval of broad-spectrum histone deacetylase (HDAC) inhibitors has firmly laid the cancer community to explore HDAC inhibition as a therapeutic approach for cancer treatment. Hitting one HDAC member could yield clinical benefit but this required a complete understanding of the functions of the different HDAC members. Here we explored the consequences of specific HDAC5 inhibition in cancer cells. We demonstrated that HDAC5 inhibition induces an iron-dependent reactive oxygen species (ROS) production, ultimately leading to apoptotic cell death as well as mechanisms of mitochondria quality control (mitophagy and mitobiogenesis). Interestingly, adaptation of HDAC5-depleted cells to oxidative stress passes through reprogramming of metabolic pathways towards glucose and glutamine. Therefore, interference with both glucose and glutamine supply in HDAC5-inhibited cancer cells significantly increases apoptotic cell death and reduces tumour growth in vivo; providing insight into a valuable clinical strategy combining the selective inhibition of HDAC5 with various inhibitors of metabolism as a new therapy to kill cancer cells.
Collapse
Affiliation(s)
- E Hendrick
- University of Liege, GIGA-Molecular Biology of Diseases, Protein Signalisation and Interaction (PSI) Laboratory, Liège, Belgium
| | - P Peixoto
- University of Liege, GIGA-Cancer, Metastasis Research Laboratory (MRL), Liège, Belgium
| | - A Blomme
- University of Liege, GIGA-Cancer, Metastasis Research Laboratory (MRL), Liège, Belgium
| | - C Polese
- University of Liege, GIGA-Molecular Biology of Diseases, Protein Signalisation and Interaction (PSI) Laboratory, Liège, Belgium
| | - N Matheus
- University of Liege, GIGA-Molecular Biology of Diseases, Protein Signalisation and Interaction (PSI) Laboratory, Liège, Belgium
| | - J Cimino
- University of Liege, GIGA-Cancer, Laboratory of Tumor and Development Biology (LBTD), Liège, Belgium
| | - A Frère
- University of Liege, GIGA-Molecular Biology of Diseases, Protein Signalisation and Interaction (PSI) Laboratory, Liège, Belgium.,University of Liege, Laboratory of Pharmaceutical Technology and Biopharmacy (LTPB), Center for Interdisciplinary Research on Medicines (CIRM), Liège, Belgium
| | - A Mouithys-Mickalad
- University of Liege, Centre for Oxygen, R&D (CORD), Institute of Chemistry, Liège, Belgium
| | - D Serteyn
- University of Liege, Centre for Oxygen, R&D (CORD), Institute of Chemistry, Liège, Belgium
| | - L Bettendorff
- University of Liege, GIGA-Signal Neurosciences, Laboratory of Pathological Aging and Epilepsy, Liège, Belgium
| | - B Elmoualij
- University of Liege, Department of Human Histology-CRPP, Liège, Belgium
| | - P De Tullio
- University of Liege, Drug Research Center, Center for Interdisciplinary Research on Medicines (CIRM), Medicinal Chemistry Department, Liège, Belgium
| | - G Eppe
- University of Liege, CART-LSM, Inorganic Analytical Chemistry, Chemistry Department, Liège, Belgium
| | - F Dequiedt
- University of Liege, GIGA-Molecular Biology of Diseases, Protein Signalisation and Interaction (PSI) Laboratory, Liège, Belgium
| | - V Castronovo
- University of Liege, GIGA-Cancer, Metastasis Research Laboratory (MRL), Liège, Belgium
| | - D Mottet
- University of Liege, GIGA-Molecular Biology of Diseases, Protein Signalisation and Interaction (PSI) Laboratory, Liège, Belgium
| |
Collapse
|
35
|
Frère A, Baroni A, Hendrick E, Delvigne AS, Orange F, Peulen O, Dakwar GR, Diricq J, Dubois P, Evrard B, Remaut K, Braeckmans K, De Smedt SC, Laloy J, Dogné JM, Feller G, Mespouille L, Mottet D, Piel G. PEGylated and Functionalized Aliphatic Polycarbonate Polyplex Nanoparticles for Intravenous Administration of HDAC5 siRNA in Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:2181-2195. [PMID: 28029254 DOI: 10.1021/acsami.6b15064] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Guanidine and morpholine functionalized aliphatic polycarbonate polymers are able to deliver efficiently histone deacetylase 5 (HDAC5) siRNA into the cytoplasm of cancer cells in vitro leading to a decrease of cell proliferation were previously developed. To allow these biodegradable and biocompatible polyplex nanoparticles to overcome the extracellular barriers and be effective in vivo after an intravenous injection, polyethylene glycol chains (PEG750 or PEG2000) were grafted on the polymer structure. These nanoparticles showed an average size of about 150 nm and a slightly positive ζ-potential with complete siRNA complexation. Behavior of PEGylated and non-PEGylated polyplexes were investigated in the presence of serum, in terms of siRNA complexation (fluorescence correlation spectroscopy), size (dynamic light scattering and single-particle tracking), interaction with proteins (isothermal titration calorimetry) and cellular uptake. Surprisingly, both PEGylated and non-PEGylated formulations presented relatively good behavior in the presence of fetal bovine serum (FBS). Hemocompatibility tests showed no effect of these polyplexes on hemolysis and coagulation. In vivo biodistribution in mice was performed and showed a better siRNA accumulation at the tumor site for PEGylated polyplexes. However, cellular uptake in protein-rich conditions showed that PEGylated polyplex lost their ability to interact with biological membranes and enter into cells, showing the importance to perform in vitro investigations in physiological conditions closed to in vivo situation. In vitro, the efficiency of PEGylated nanoparticles decreases compared to non-PEGylated particles, leading to the loss of the antiproliferative effect on cancer cells.
Collapse
Affiliation(s)
- Antoine Frère
- Laboratory of Pharmaceutical Technology and Biopharmacy (LTPB) - Center for Interdisciplinary Research on Medicines (CIRM), University of Liege , Avenue Hippocrate 15, 4000 Liege, Belgium
- Protein Signalisation and Interaction (PSI) - GIGA, University of Liege , Avenue de l'Hopital 11, 4000 Liege, Belgium
| | - Alexandra Baroni
- Laboratory of Polymeric and Composite Materials, Center of Innovation and Research in Materials and Polymers (CIRMAP), Research Institute for Health Sciences and Technology, University of Mons , Place du Parc 20, 7000 Mons, Belgium
| | - Elodie Hendrick
- Protein Signalisation and Interaction (PSI) - GIGA, University of Liege , Avenue de l'Hopital 11, 4000 Liege, Belgium
| | - Anne-Sophie Delvigne
- Namur Nanosafety Center (NNC), NAmur Research Institute for LIfe Sciences (NARILIS), Department of Pharmacy, University of Namur , Rue de Bruxelles 61, 5000 Namur, Belgium
| | - François Orange
- Centre Commun de Microscopie Appliquée, University of Nice-Sophia Antipolis , Parc Valrose, 06108 Nice, France
| | - Olivier Peulen
- Metastasis Research Laboratory (MRL) - GIGA, University of Liege , Avenue Hippocrate 15, 4000 Liege, Belgium
| | - George R Dakwar
- Laboratory for General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University , Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Jérôme Diricq
- Laboratory of Polymeric and Composite Materials, Center of Innovation and Research in Materials and Polymers (CIRMAP), Research Institute for Health Sciences and Technology, University of Mons , Place du Parc 20, 7000 Mons, Belgium
| | - Philippe Dubois
- Laboratory of Polymeric and Composite Materials, Center of Innovation and Research in Materials and Polymers (CIRMAP), Research Institute for Health Sciences and Technology, University of Mons , Place du Parc 20, 7000 Mons, Belgium
| | - Brigitte Evrard
- Laboratory of Pharmaceutical Technology and Biopharmacy (LTPB) - Center for Interdisciplinary Research on Medicines (CIRM), University of Liege , Avenue Hippocrate 15, 4000 Liege, Belgium
| | - Katrien Remaut
- Laboratory for General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University , Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory for General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University , Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory for General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University , Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Julie Laloy
- Namur Nanosafety Center (NNC), NAmur Research Institute for LIfe Sciences (NARILIS), Department of Pharmacy, University of Namur , Rue de Bruxelles 61, 5000 Namur, Belgium
| | - Jean-Michel Dogné
- Namur Nanosafety Center (NNC), NAmur Research Institute for LIfe Sciences (NARILIS), Department of Pharmacy, University of Namur , Rue de Bruxelles 61, 5000 Namur, Belgium
| | - Georges Feller
- Laboratory of Biochemistry, Centre for Protein Engineering (CIP), University of Liège , Allée du 6 Août 13, 4000 Liège, Belgium
| | - Laetitia Mespouille
- Laboratory of Polymeric and Composite Materials, Center of Innovation and Research in Materials and Polymers (CIRMAP), Research Institute for Health Sciences and Technology, University of Mons , Place du Parc 20, 7000 Mons, Belgium
| | - Denis Mottet
- Protein Signalisation and Interaction (PSI) - GIGA, University of Liege , Avenue de l'Hopital 11, 4000 Liege, Belgium
| | - Géraldine Piel
- Laboratory of Pharmaceutical Technology and Biopharmacy (LTPB) - Center for Interdisciplinary Research on Medicines (CIRM), University of Liege , Avenue Hippocrate 15, 4000 Liege, Belgium
| |
Collapse
|
36
|
López JE, Sullivan ED, Fierke CA. Metal-dependent Deacetylases: Cancer and Epigenetic Regulators. ACS Chem Biol 2016; 11:706-16. [PMID: 26907466 DOI: 10.1021/acschembio.5b01067] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Epigenetic regulation is a key factor in cellular homeostasis. Post-translational modifications (PTMs) are a central focus of this regulation as they function as signaling markers within the cell. Lysine acetylation is a dynamic, reversible PTM that has garnered recent attention due to alterations in various types of cancer. Acetylation levels are regulated by two opposing enzyme families: lysine acetyltransferases (KATs) and histone deacetylases (HDACs). HDACs are key players in epigenetic regulation and have a role in the silencing of tumor suppressor genes. The dynamic equilibrium of acetylation makes HDACs attractive targets for drug therapy. However, substrate selectivity and biological function of HDAC isozymes is poorly understood. This review outlines the current understanding of the roles and specific epigenetic interactions of the metal-dependent HDACs in addition to their roles in cancer.
Collapse
Affiliation(s)
- Jeffrey E. López
- Interdepartmental
Program in Chemical Biology, University of Michigan, 210 Washtenaw
Avenue, Ann Arbor, Michigan 48109-2216, United States
| | - Eric D. Sullivan
- Interdepartmental
Program in Chemical Biology, University of Michigan, 210 Washtenaw
Avenue, Ann Arbor, Michigan 48109-2216, United States
| | - Carol A. Fierke
- Interdepartmental
Program in Chemical Biology, University of Michigan, 210 Washtenaw
Avenue, Ann Arbor, Michigan 48109-2216, United States
- Departments
of Chemistry and Biological Chemistry, University of Michigan, 930 North
University Avenue, Ann Arbor, Michigan 48109-2216, United States
| |
Collapse
|
37
|
Cao X, Liu DH, Zhou Y, Yan XM, Yuan LQ, Pan J, Fu MC, Zhang T, Wang J. Histone deacetylase 5 promotes Wilms' tumor cell proliferation through the upregulation of c-Met. Mol Med Rep 2016; 13:2745-50. [PMID: 26847592 DOI: 10.3892/mmr.2016.4828] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 06/30/2015] [Indexed: 11/06/2022] Open
Abstract
The histone deacetylase (HDAC) family is comprised of enzymes, which are involved in modulating the majority of critical cellular processes, including transcriptional regulation, apoptosis, proliferation and cell cycle progression. However, the biological function of HDAC5 in Wilms' tumor remains to be fully elucidated. The present study aimed to investigate the expression and function of HDAC5 in Wilm's tumor. It was demonstrated that the mRNA and protein levels of HDAC5 were upregulated in human Wilms' tumor tissues. Overexpression of HDAC5 in G401 cells was observed to significantly promote cellular proliferation, as demonstrated by the results of an MTT assay and bromodeoxyuridine incorporation assay. By contrast, HDAC5 knockdown using small interfering RNA suppressed the proliferation of the G401 cells. At the molecular level, the present study demonstrated that HDAC5 promoted the expression of c‑Met, which has been previously identified as an oncogene. In addition, downregulation of c‑Met inhibited the proliferative effects of HDAC5 in human Wilms' tumor cells. Taken together, these results suggested that HDAC5 promotes cellular proliferation through the upregulation of c‑Met, and may provide a novel therapeutic target for the treatment of patients with Wilms' tumor.
Collapse
Affiliation(s)
- Xu Cao
- Department of Surgery, Childrens' Hospital Affiliated to Soochow University, Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - De-Hong Liu
- Department of Pediatric Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Yun Zhou
- Department of Surgery, Childrens' Hospital Affiliated to Soochow University, Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Xiang-Ming Yan
- Department of Surgery, Childrens' Hospital Affiliated to Soochow University, Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Li-Qun Yuan
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Jian Pan
- Department of Surgery, Childrens' Hospital Affiliated to Soochow University, Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Ming-Cui Fu
- Department of Surgery, Childrens' Hospital Affiliated to Soochow University, Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Ting Zhang
- Department of Surgery, Childrens' Hospital Affiliated to Soochow University, Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Jian Wang
- Department of Surgery, Childrens' Hospital Affiliated to Soochow University, Soochow University, Suzhou, Jiangsu 215003, P.R. China
| |
Collapse
|
38
|
Fahmy K, Gonzalez A, Arafa M, Peixoto P, Bellahcène A, Turtoi A, Delvenne P, Thiry M, Castronovo V, Peulen O. Myoferlin plays a key role in VEGFA secretion and impacts tumor-associated angiogenesis in human pancreas cancer. Int J Cancer 2015; 138:652-63. [PMID: 26311411 DOI: 10.1002/ijc.29820] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 07/31/2015] [Accepted: 08/17/2015] [Indexed: 01/14/2023]
Abstract
Pancreatic ductal adenocarcinoma is one of the most deadly forms of cancers with no satisfactory treatment to date. Recent studies have identified myoferlin, a ferlin family member, in human pancreas adenocarcinoma where its expression was associated to a bad prognosis. However, the function of myoferlin in pancreas adenocarcinoma has not been reported. In other cell types, myoferlin is involved in several key plasma membrane processes such as fusion, repair, endocytosis and tyrosine kinase receptor activity. In this study, we showed that myoferlin silencing in BxPC-3 human pancreatic cancer cells resulted in the inhibition of cell proliferation in vitro and in a significant reduction of the tumor volume in chick chorioallantoic membrane assay. In addition to be smaller, the tumors formed by the myoferlin-silenced cells showed a marked absence of functional blood vessels. We further demonstrated that this effect was due, at least in part, to an inhibition of VEGFA secretion by BxPC-3 myoferlin-silenced cells. Using immunofluorescence and electron microscopy, we linked the decreased VEGFA secretion to an impairment of VEGFA exocytosis. The clinical relevance of our results was further strengthened by a significant correlation between myoferlin expression in a series of human pancreatic malignant lesions and their angiogenic status evaluated by the determination of the blood vessel density.
Collapse
Affiliation(s)
- Karim Fahmy
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Arnaud Gonzalez
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Mohammad Arafa
- Department of Pathology, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
| | - Paul Peixoto
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Andrei Turtoi
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Philippe Delvenne
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Marc Thiry
- Laboratory of Cell Biology, GIGA-R, University of Liege, Liege, Belgium
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, Liege, Belgium
| |
Collapse
|
39
|
Liu Q, Sun Y, Zheng JM, Yan XL, Chen HM, Chen JK, Huang HQ. Formononetin sensitizes glioma cells to doxorubicin through preventing EMT via inhibition of histone deacetylase 5. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:6434-6441. [PMID: 26261519 PMCID: PMC4525853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 03/30/2015] [Indexed: 06/04/2023]
Abstract
Chemoresistance is a major obstacle to successful chemotherapy for glioma. Formononetin is a novel herbal isoflavonoid isolated from Astragalus membranaceus and possesses antitumorigenic properties. In the present study, we investigated the anti-proliferative effects of formononetin on human glioma cells, and further elucidated the molecular mechanism underlying the anti-tumor property. We found that formononetin enhanced doxorubicin cytotoxicity in glioma cells. Combined treatment with formononetin reversed the doxorubicin-induced epithelial-mesenchymal transition (EMT) in tumor cells. Moreover, we found that formononetin treatment significantly decreased the expression of HDAC5. Overexpression of HDAC5 diminished the suppressive effects of formononetin on glioma cell viability. Furthermore, knockdown of HDAC5 by siRNA inhibited the doxorubicin-induced EMT in glioma cells. Taken together, these results demonstrated that formononetin-combined therapy may enhance the therapeutic efficacy of doxorubicin in glioma cells by preventing EMT through inhibition of HDAC5.
Collapse
Affiliation(s)
- Quan Liu
- Department of Neurosurgery, The Fourth Affiliated Hospital Guangxi Medical UniversityLiuzhou 545005, Guangxi, China
| | - Yan Sun
- Department of Scientific Research and Education, The Fourth Affiliated Hospital Guangxi Medical UniversityLiuzhou 545005, Guangxi, China
| | - Jie-Min Zheng
- Department of Neurosurgery, The Fourth Affiliated Hospital Guangxi Medical UniversityLiuzhou 545005, Guangxi, China
| | - Xian-Lei Yan
- Department of Neurosurgery, The Fourth Affiliated Hospital Guangxi Medical UniversityLiuzhou 545005, Guangxi, China
| | - Hong-Mou Chen
- Department of Neurosurgery, The Fourth Affiliated Hospital Guangxi Medical UniversityLiuzhou 545005, Guangxi, China
| | - Jia-Kang Chen
- Department of Neurosurgery, The Fourth Affiliated Hospital Guangxi Medical UniversityLiuzhou 545005, Guangxi, China
| | - He-Qing Huang
- Department of Neurosurgery, The Fourth Affiliated Hospital Guangxi Medical UniversityLiuzhou 545005, Guangxi, China
| |
Collapse
|
40
|
Sui X, Zhu J, Zhou J, Wang X, Li D, Han W, Fang Y, Pan H. Epigenetic modifications as regulatory elements of autophagy in cancer. Cancer Lett 2015; 360:106-13. [DOI: 10.1016/j.canlet.2015.02.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 02/04/2015] [Accepted: 02/08/2015] [Indexed: 12/16/2022]
|
41
|
HDAC Family Members Intertwined in the Regulation of Autophagy: A Druggable Vulnerability in Aggressive Tumor Entities. Cells 2015; 4:135-68. [PMID: 25915736 PMCID: PMC4493453 DOI: 10.3390/cells4020135] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/15/2015] [Accepted: 04/15/2015] [Indexed: 12/21/2022] Open
Abstract
The exploitation of autophagy by some cancer entities to support survival and dodge death has been well-described. Though its role as a constitutive process is important in normal, healthy cells, in the milieu of malignantly transformed and highly proliferative cells, autophagy is critical for escaping metabolic and genetic stressors. In recent years, the importance of histone deacetylases (HDACs) in cancer biology has been heavily investigated, and the enzyme family has been shown to play a role in autophagy, too. HDAC inhibitors (HDACi) are being integrated into cancer therapy and clinical trials are ongoing. The effect of HDACi on autophagy and, conversely, the effect of autophagy on HDACi efficacy are currently under investigation. With the development of HDACi that are able to selectively target individual HDAC isozymes, there is great potential for specific therapy that has more well-defined effects on cancer biology and also minimizes toxicity. Here, the role of autophagy in the context of cancer and the interplay of this process with HDACs will be summarized. Identification of key HDAC isozymes involved in autophagy and the ability to target specific isozymes yields the potential to cripple and ultimately eliminate malignant cells depending on autophagy as a survival mechanism.
Collapse
|
42
|
Frère A, Kawalec M, Tempelaar S, Peixoto P, Hendrick E, Peulen O, Evrard B, Dubois P, Mespouille L, Mottet D, Piel G. Impact of the Structure of Biocompatible Aliphatic Polycarbonates on siRNA Transfection Ability. Biomacromolecules 2015; 16:769-79. [DOI: 10.1021/bm501676p] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
| | - Michal Kawalec
- Laboratory
of Polymeric and Composite Materials, Center of Innovation and Research
in Materials and Polymers (CIRMAP), Research Institute for Health
Sciences and Technology, University of Mons, Place du Parc 20 - 7000, Mons, Belgium
| | - Sarah Tempelaar
- Laboratory
of Polymeric and Composite Materials, Center of Innovation and Research
in Materials and Polymers (CIRMAP), Research Institute for Health
Sciences and Technology, University of Mons, Place du Parc 20 - 7000, Mons, Belgium
| | | | | | | | | | - Philippe Dubois
- Laboratory
of Polymeric and Composite Materials, Center of Innovation and Research
in Materials and Polymers (CIRMAP), Research Institute for Health
Sciences and Technology, University of Mons, Place du Parc 20 - 7000, Mons, Belgium
| | - Laetitia Mespouille
- Laboratory
of Polymeric and Composite Materials, Center of Innovation and Research
in Materials and Polymers (CIRMAP), Research Institute for Health
Sciences and Technology, University of Mons, Place du Parc 20 - 7000, Mons, Belgium
| | | | | |
Collapse
|
43
|
Thaler F, Mercurio C. Towards selective inhibition of histone deacetylase isoforms: what has been achieved, where we are and what will be next. ChemMedChem 2014; 9:523-6. [PMID: 24730063 DOI: 10.1002/cmdc.201300413] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Histone deacetylases (HDACs) are widely studied targets for the treatment of cancer and other diseases. Up to now, over twenty HDAC inhibitors have entered clinical studies and two of them have already reached the market, namely the hydroxamic acid derivative SAHA (vorinostat, Zolinza) and the cyclic depsipeptide FK228 (romidepsin, Istodax) that have been approved for the treatment of cutaneous T-cell lymphoma (CTCL). A common aspect of the first HDAC inhibitors is the absence of any particular selectivity towards specific isozymes. Some of molecules resulted to be “pan”-HDAC inhibitors, while others are class I selective. In the meantime, the knowledge of HDAC biology has continuously progressed. Key advances in the structural biology of various isozymes, reliable molecular homology models as well as suitable biological assays have provided new tools for drug discovery activities. This Minireview aims at surveying these recent developments as well as the design, synthesis and biological characterization of isoform-selective derivatives.
Collapse
|
44
|
Fan J, Lou B, Chen W, Zhang J, Lin S, Lv FF, Chen Y. Down-regulation of HDAC5 inhibits growth of human hepatocellular carcinoma by induction of apoptosis and cell cycle arrest. Tumour Biol 2014; 35:11523-11532. [PMID: 25129440 DOI: 10.1007/s13277-014-2358-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/14/2014] [Indexed: 10/24/2022] Open
Abstract
Histone deacetylases (HDACs) play a critical role in the proliferation, differentiation, and apoptosis of cancer cells. An obstacle for the application of HDAC inhibitors as effective anti-cancer therapeutics is that our current knowledge on the contributions of different HDACs in various cancer types remains scarce. The present study reported that the mRNA and protein levels of HDAC5 were up-regulated in human hepatocellular carcinoma (HCC) tissues and cells as shown by quantitative real-time PCR and Western blot. MTT assay and BrdU incorporation assay showed that the down-regulation of HDAC5 inhibited cell proliferation in HepG2, Hep3B, and Huh7 cell lines. Data from in vivo xenograft tumorigenesis model also demonstrated the anti-proliferative effect of HDAC5 depletion on tumor cell growth. Furthermore, the suppression of HDAC5 promoted cell apoptosis and induced G1-phase cell cycle arrest in HCC cells. On the molecular level, we observed altered expression of apoptosis-related proteins such as p53, bax, bcl-2, cyto C, and caspase 3 in HDAC5-shRNA-transfected cells. Knockdown of HDAC5 led to a significant up-regulation of p21 and down-regulation of cyclin D1 and CDK2/4/6. We also found that the down-regulation of HDAC5 substantially increased p53 stability and promoted its nuclear localization and transcriptional activity. Our study suggested that knockdown of HDAC5 could inhibit cancer cell proliferation by the induction of cell cycle arrest and apoptosis; thus, suppression of HDAC5 may be a viable option for treating HCC patients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Carcinoma, Hepatocellular/enzymology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Cell Cycle
- Cell Proliferation
- Female
- Gene Expression Regulation, Neoplastic
- Histone Deacetylases/chemistry
- Histone Deacetylases/genetics
- Histone Deacetylases/metabolism
- Humans
- Liver Neoplasms/enzymology
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Luciferases/metabolism
- Male
- Mice
- Mice, Nude
- Middle Aged
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jian Fan
- Department of Laboratory Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79, Qingchun Road, Hangzhou, Zhejiang Province, 310003, China
| | | | | | | | | | | | | |
Collapse
|
45
|
Polese C, Mottet D. [HDAC5 inhibition: a tool to stop cancer cell immortality]. Med Sci (Paris) 2014; 30:730-2. [PMID: 25174744 DOI: 10.1051/medsci/20143008005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Catherine Polese
- Laboratoire de recherche sur les métastases, groupe HDAC, Université de Liège, Institut de pathologie B23, B-4000 Liège (Sart-Tilman), Belgique - Adresse actuelle : Signalisation et interaction des protéines, GIGA-R (B34), avenue de l'Hôpital, 1, B-4000 Liège, Belgique
| | - Denis Mottet
- Laboratoire de recherche sur les métastases, groupe HDAC, Université de Liège, Institut de pathologie B23, B-4000 Liège (Sart-Tilman), Belgique - Adresse actuelle : Signalisation et interaction des protéines, GIGA-R (B34), avenue de l'Hôpital, 1, B-4000 Liège, Belgique
| |
Collapse
|
46
|
Sun Y, Liu PY, Scarlett CJ, Malyukova A, Liu B, Marshall GM, MacKenzie KL, Biankin AV, Liu T. Histone deacetylase 5 blocks neuroblastoma cell differentiation by interacting with N-Myc. Oncogene 2014; 33:2987-94. [PMID: 23812427 DOI: 10.1038/onc.2013.253] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 05/10/2013] [Accepted: 05/31/2013] [Indexed: 12/18/2022]
Abstract
The N-Myc oncoprotein induces neuroblastoma, which arises from undifferentiated neuroblasts in the sympathetic nervous system, by modulating gene and protein expression and consequently causing cell differentiation block and cell proliferation. The class IIa histone deacetylase 5 (HDAC5) represses gene transcription, and blocks myoblast, osteoblast and leukemia cell differentiation. Here we showed that N-Myc upregulated HDAC5 expression in neuroblastoma cells. Conversely, HDAC5 repressed the ubiquitin-protein ligase NEDD4 gene expression, increased Aurora A gene expression and consequently upregulated N-Myc protein expression. Genome-wide gene expression analysis and protein co-immunoprecipitation assays revealed that HDAC5 and N-Myc repressed the expression of a common subset of genes by forming a protein complex, whereas HDAC5 and the class III HDAC SIRT2 independently repressed the expression of another common subset of genes without forming a protein complex. Moreover, HDAC5 blocked differentiation and induced proliferation in neuroblastoma cells. Taken together, our data identify HDAC5 as a novel co-factor in N-Myc oncogenesis, and provide the evidence for the potential application of HDAC5 inhibitors in the therapy of N-Myc-induced neuroblastoma and potentially other c-Myc-induced malignancies.
Collapse
Affiliation(s)
- Y Sun
- Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia
| | - P Y Liu
- Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia
| | - C J Scarlett
- 1] School of Environmental and Life Sciences, University of Newcastle, Ourimbah, New South Wales, Australia [2] Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - A Malyukova
- Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia
| | - B Liu
- 1] Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia [2] Kids Cancer Alliance, Randwick, New South Wales, Australia
| | - G M Marshall
- 1] Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia [2] Kids Cancer Centre, Sydney Children's Hospital, Randwick, New South Wales, Australia
| | - K L MacKenzie
- Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia
| | - A V Biankin
- 1] Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia [2] Department of Surgery, Bankstown Hospital, Bankstown, New South Wales, Australia [3] South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Liverpool, New South Wales, Australia
| | - T Liu
- 1] Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia [2] School of Women's and Children's Health, UNSW Medicine, University of New South Wales, Randwick, New South Wales, Australia
| |
Collapse
|
47
|
Park SH, Yu SE, Chai YG, Jang YK. CDK2-dependent phosphorylation of Suv39H1 is involved in control of heterochromatin replication during cell cycle progression. Nucleic Acids Res 2014; 42:6196-207. [PMID: 24728993 PMCID: PMC4041437 DOI: 10.1093/nar/gku263] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 02/26/2014] [Accepted: 03/19/2014] [Indexed: 01/24/2023] Open
Abstract
Although several studies have suggested that the functions of heterochromatin regulators may be regulated by post-translational modifications during cell cycle progression, regulation of the histone methyltransferase Suv39H1 is not fully understood. Here, we demonstrate a direct link between Suv39H1 phosphorylation and cell cycle progression. We show that CDK2 phosphorylates Suv39H1 at Ser391 and these phosphorylation levels oscillate during the cell cycle, peaking at S phase and maintained during S-G2-M phase. The CDK2-mediated phosphorylation of Suv39H1 at Ser391 results in preferential dissociation from chromatin. Furthermore, phosphorylation-mediated dissociation of Suv39H1 from chromatin causes an enhanced occupancy of JMJD2A histone demethylase on heterochromatin and alterations in inactive histone marks. Overexpression of phospho-mimic Suv39H1 induces early replication of heterochromatin, suggesting the importance of Suv39H1 phosphorylation in the replication of heterochromatin. Moreover, overexpression of phospho-defective Suv39H1 caused altered replication timing of heterochromatin and increases sensitivity to replication stress. Collectively, our data suggest that phosphorylation-mediated modulation of Suv39H1-chromatin association may be an initial step in heterochromatin replication.
Collapse
Affiliation(s)
- Su Hyung Park
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea Initiative for Biological Function & Systems, Yonsei University, Seoul 120-749, Republic of Korea
| | - Seung Eun Yu
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea Initiative for Biological Function & Systems, Yonsei University, Seoul 120-749, Republic of Korea
| | - Young Gyu Chai
- Division of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea
| | - Yeun Kyu Jang
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea Initiative for Biological Function & Systems, Yonsei University, Seoul 120-749, Republic of Korea
| |
Collapse
|
48
|
Parbin S, Kar S, Shilpi A, Sengupta D, Deb M, Rath SK, Patra SK. Histone deacetylases: a saga of perturbed acetylation homeostasis in cancer. J Histochem Cytochem 2014; 62:11-33. [PMID: 24051359 PMCID: PMC3873803 DOI: 10.1369/0022155413506582] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the current era of genomic medicine, diseases are identified as manifestations of anomalous patterns of gene expression. Cancer is the principal example among such maladies. Although remarkable progress has been achieved in the understanding of the molecular mechanisms involved in the genesis and progression of cancer, its epigenetic regulation, particularly histone deacetylation, demands further studies. Histone deacetylases (HDACs) are one of the key players in the gene expression regulation network in cancer because of their repressive role on tumor suppressor genes. Higher expression and function of deacetylases disrupt the finely tuned acetylation homeostasis in both histone and non-histone target proteins. This brings about alterations in the genes implicated in the regulation of cell proliferation, differentiation, apoptosis and other cellular processes. Moreover, the reversible nature of epigenetic modulation by HDACs makes them attractive targets for cancer remedy. This review summarizes the current knowledge of HDACs in tumorigenesis and tumor progression as well as their contribution to the hallmarks of cancer. The present report also describes briefly various assays to detect histone deacetylase activity and discusses the potential role of histone deacetylase inhibitors as emerging epigenetic drugs to cure cancer.
Collapse
Affiliation(s)
- Sabnam Parbin
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, India (SP, SK, AS, DS, SKR, SKP)
| | | | | | | | | | | | | |
Collapse
|
49
|
Stat3 inhibits PTPN13 expression in squamous cell lung carcinoma through recruitment of HDAC5. BIOMED RESEARCH INTERNATIONAL 2013; 2013:468963. [PMID: 24191246 PMCID: PMC3804148 DOI: 10.1155/2013/468963] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 08/17/2013] [Accepted: 08/30/2013] [Indexed: 01/05/2023]
Abstract
Proteins of the protein tyrosine phosphatase (PTP) family are known to be signaling molecules that regulate a variety of cellular processes including cell growth, differentiation, and apoptosis. PTPN13 (also known as FAP1, PTPL1, PTPLE, PTPBAS, and PTP1E), a putative tumor suppressor, is frequently inactivated in lung carcinoma through the loss of either mRNA or protein expression. However, the molecular mechanisms underlying its dysregulation have not been fully explored. Interleukin-6 (IL-6) mediated Stat3 activation is viewed as crucial for multiple tumor growth and progression. Here, we demonstrate that PTPN13 is a direct transcriptional target of Stat3 in the squamous cell lung carcinoma. Our data show that IL-6 administration or transfection of a constitutively activated Stat3 in HCC-1588 and SK-MES-1 cells inhibits PTPN13 mRNA transcription. Using luciferase reporter and ChIP assays, we show that Stat3 binds to the promoter region of PTPN13 and promotes its activity through recruiting HDAC5. Thus, our results suggest a previously unknown Stat3-PTPN13 molecular network controlling squamous cell lung carcinoma development.
Collapse
|
50
|
The anti-tumor effect of HDAC inhibition in a human pancreas cancer model is significantly improved by the simultaneous inhibition of cyclooxygenase 2. PLoS One 2013; 8:e75102. [PMID: 24040391 PMCID: PMC3770617 DOI: 10.1371/journal.pone.0075102] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 08/12/2013] [Indexed: 12/30/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is the fourth leading cause of cancer death worldwide, with no satisfactory treatment to date. In this study, we tested whether the combined inhibition of cyclooxygenase-2 (COX-2) and class I histone deacetylase (HDAC) may results in a better control of pancreatic ductal adenocarcinoma. The impact of the concomitant HDAC and COX-2 inhibition on cell growth, apoptosis and cell cycle was assessed first in vitro on human pancreas BxPC-3, PANC-1 or CFPAC-1 cells treated with chemical inhibitors (SAHA, MS-275 and celecoxib) or HDAC1/2/3/7 siRNA. To test the potential antitumoral activity of this combination in vivo, we have developed and characterized, a refined chick chorioallantoic membrane tumor model that histologically and proteomically mimics human pancreatic ductal adenocarcinoma. The combination of HDAC1/3 and COX-2 inhibition significantly impaired proliferation of BxPC-3 cells in vitro and stalled entirely the BxPC-3 cells tumor growth onto the chorioallantoic membrane in vivo. The combination was more effective than either drug used alone. Consistently, we showed that both HDAC1 and HDAC3 inhibition induced the expression of COX-2 via the NF-kB pathway. Our data demonstrate, for the first time in a Pancreatic Ductal Adenocarcinoma (PDAC) model, a significant action of HDAC and COX-2 inhibitors on cancer cell growth, which sets the basis for the development of potentially effective new combinatory therapies for pancreatic ductal adenocarcinoma patients.
Collapse
|