1
|
Della Via FI, Alvarez MC, Basting RT, Saad STO. The Effects of Green Tea Catechins in Hematological Malignancies. Pharmaceuticals (Basel) 2023; 16:1021. [PMID: 37513933 PMCID: PMC10385775 DOI: 10.3390/ph16071021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Green tea catechins are bioactive polyphenol compounds which have attracted significant attention for their diverse biological activities and potential health benefits. Notably, epigallocatechin-3-gallate (EGCG) has emerged as a potent apoptosis inducer through mechanisms involving caspase activation, modulation of Bcl-2 family proteins, disruption of survival signaling pathways and by regulating the redox balance, inducing oxidative stress. Furthermore, emerging evidence suggests that green tea catechins can modulate epigenetic alterations, including DNA methylation and histone modifications. In addition to their apoptotic actions, ROS signaling effects and reversal of epigenetic alterations, green tea catechins have shown promising results in promoting the differentiation of leukemia cells. This review highlights the comprehensive actions of green tea catechins and provides valuable insights from clinical trials investigating the therapeutic potential of green tea catechins in leukemia treatment. Understanding these multifaceted mechanisms and the outcomes of clinical trials may pave the way for the development of innovative strategies and the integration of green tea catechins into clinical practice for improving leukemia patient outcomes.
Collapse
Affiliation(s)
- Fernanda Isabel Della Via
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro, UNICAMP, Rua Carlos Chagas 480, Campinas 13083-878, SP, Brazil
| | - Marisa Claudia Alvarez
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro, UNICAMP, Rua Carlos Chagas 480, Campinas 13083-878, SP, Brazil
| | - Rosanna Tarkany Basting
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro, UNICAMP, Rua Carlos Chagas 480, Campinas 13083-878, SP, Brazil
| | - Sara Teresinha Olalla Saad
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro, UNICAMP, Rua Carlos Chagas 480, Campinas 13083-878, SP, Brazil
| |
Collapse
|
2
|
Otsuka K, Iwasaki T. Insights into radiation carcinogenesis based on dose-rate effects in tissue stem cells. Int J Radiat Biol 2023; 99:1503-1521. [PMID: 36971595 DOI: 10.1080/09553002.2023.2194398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 03/16/2023] [Indexed: 03/29/2023]
Abstract
PURPOSE Increasing epidemiological and biological evidence suggests that radiation exposure enhances cancer risk in a dose-dependent manner. This can be attributed to the 'dose-rate effect,' where the biological effect of low dose-rate radiation is lower than that of the same dose at a high dose-rate. This effect has been reported in epidemiological studies and experimental biology, although the underlying biological mechanisms are not completely understood. In this review, we aim to propose a suitable model for radiation carcinogenesis based on the dose-rate effect in tissue stem cells. METHODS We surveyed and summarized the latest studies on the mechanisms of carcinogenesis. Next, we summarized the radiosensitivity of intestinal stem cells and the role of dose-rate in the modulation of stem-cell dynamics after irradiation. RESULTS Consistently, driver mutations can be detected in most cancers from past to present, supporting the hypothesis that cancer progression is initiated by the accumulation of driver mutations. Recent reports demonstrated that driver mutations can be observed even in normal tissues, which suggests that the accumulation of mutations is a necessary condition for cancer progression. In addition, driver mutations in tissue stem cells can cause tumors, whereas they are not sufficient when they occur in non-stem cells. For non-stem cells, tissue remodeling induced by marked inflammation after the loss of tissue cells is important in addition to the accumulation of mutations. Therefore, the mechanism of carcinogenesis differs according to the cell type and magnitude of stress. In addition, our results indicated that non-irradiated stem cells tend to be eliminated from three-dimensional cultures of intestinal stem cells (organoids) composed of irradiated and non-irradiated stem cells, supporting the stem-cell competition. CONCLUSIONS We propose a unique scheme in which the dose-rate dependent response of intestinal stem cells incorporates the concept of the threshold of stem-cell competition and context-dependent target shift from stem cells to whole tissue. The concept highlights four key issues that should be considered in radiation carcinogenesis: i.e. accumulation of mutations; tissue reconstitution; stem-cell competition; and environmental factors like epigenetic modifications.
Collapse
Affiliation(s)
- Kensuke Otsuka
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry, Tokyo, Japan
| | - Toshiyasu Iwasaki
- Strategy and Planning Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry, Tokyo, Japan
| |
Collapse
|
3
|
Genetic and Epigenetic Aspects of Atopic Dermatitis. Int J Mol Sci 2020; 21:ijms21186484. [PMID: 32899887 PMCID: PMC7554821 DOI: 10.3390/ijms21186484] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022] Open
Abstract
Atopic dermatitis is a heterogeneous disease, in which the pathogenesis is associated with mutations in genes encoding epidermal structural proteins, barrier enzymes, and their inhibitors; the role of genes regulating innate and adaptive immune responses and environmental factors inducing the disease is also noted. Recent studies point to the key role of epigenetic changes in the development of the disease. Epigenetic modifications are mainly mediated by DNA methylation, histone acetylation, and the action of specific non-coding RNAs. It has been documented that the profile of epigenetic changes in patients with atopic dermatitis (AD) differs from that observed in healthy people. This applies to the genes affecting the regulation of immune response and inflammatory processes, e.g., both affecting Th1 bias and promoting Th2 responses and the genes of innate immunity, as well as those encoding the structural proteins of the epidermis. Understanding of the epigenetic alterations is therefore pivotal to both create new molecular classifications of atopic dermatitis and to enable the development of personalized treatment strategies.
Collapse
|
4
|
Ionizing Radiation-Induced Epigenetic Modifications and Their Relevance to Radiation Protection. Int J Mol Sci 2020; 21:ijms21175993. [PMID: 32825382 PMCID: PMC7503247 DOI: 10.3390/ijms21175993] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
The present system of radiation protection assumes that exposure at low doses and/or low dose-rates leads to health risks linearly related to the dose. They are evaluated by a combination of epidemiological data and radiobiological models. The latter imply that radiation induces deleterious effects via genetic mutation caused by DNA damage with a linear dose-dependence. This picture is challenged by the observation of radiation-induced epigenetic effects (changes in gene expression without altering the DNA sequence) and of non-linear responses, such as non-targeted and adaptive responses, that in turn can be controlled by gene expression networks. Here, we review important aspects of the biological response to ionizing radiation in which epigenetic mechanisms are, or could be, involved, focusing on the possible implications to the low dose issue in radiation protection. We examine in particular radiation-induced cancer, non-cancer diseases and transgenerational (hereditary) effects. We conclude that more realistic models of radiation-induced cancer should include epigenetic contribution, particularly in the initiation and progression phases, while the impact on hereditary risk evaluation is expected to be low. Epigenetic effects are also relevant in the dispute about possible "beneficial" effects at low dose and/or low dose-rate exposures, including those given by the natural background radiation.
Collapse
|
5
|
Gao C, Zhuang J, Zhou C, Liu L, Liu C, Li H, Zhao M, Liu G, Sun C. Developing DNA methylation-based prognostic biomarkers of acute myeloid leukemia. J Cell Biochem 2018; 119:10041-10050. [PMID: 30171717 DOI: 10.1002/jcb.27336] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/26/2018] [Indexed: 12/21/2022]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous clonal neoplasm characterized by complex genomic alterations. The incidence of AML increases with age, and most cases experience serious illness and poor prognosis. To explore the relationship between abnormal DNA methylation and the occurrence and development of AML based on the Gene Expression Database (GEO), this study extracted data related to methylation in AML and identified a methylated CpG site that was significantly different in terms of expression and distribution between the primary cells of AML patients, and hematopoietic stem/progenitor cells from normal bone marrow. To further investigate the differences caused by the dysfunction of methylation sites, bioinformatics analysis was used to screen methylation-related biomarkers, and the potential prognostic genes were selected by univariate and multivariate Cox proportional hazards regressions. Finally, five independent prognostic indicators were identified. In addition, these results provide new insight into the molecular mechanisms of methylation.
Collapse
Affiliation(s)
- Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, China.,Department of Oncology, Affilited Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Chao Zhou
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, China.,Department of Oncology, Affilited Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Lijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, China.,Department of Oncology, Affilited Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Huayao Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Minzhang Zhao
- School of Medicine, Shandong University, Jinan, China
| | - Gongxi Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, China.,Department of Oncology, Affilited Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, China.,Department of Oncology, Affilited Hospital of Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
6
|
The natural phenolic peperobtusin A induces apoptosis of lymphoma U937 cells via the Caspase dependent and p38 MAPK signaling pathways. Biomed Pharmacother 2018; 102:772-781. [DOI: 10.1016/j.biopha.2018.03.141] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 11/20/2022] Open
|
7
|
Nikoonahad Lotfabadi N, Mohseni Kouchesfahani H, Sheikhha MH, Kalantar SM. In vitro transfection of anti-tumor miR-101 induces BIM, a pro-apoptotic protein, expression in acute myeloid leukemia (AML). EXCLI JOURNAL 2017; 16:1257-1267. [PMID: 29333128 PMCID: PMC5763080 DOI: 10.17179/excli2017-721] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 11/11/2017] [Indexed: 12/14/2022]
Abstract
Acute myeloid leukemia (AML) frequently relapses after initial treatment, though it is possible that drug resistance occurs. Hence, it seems necessary to develop novel therapies such as gene therapy specifically via miRNA transfection. MicroRNA-101 has been considered as a tumor suppressor in different types of cancer. It is demonstrated that exogenous miR-101 transfection is associated with decreased viability in AML in this paper. Besides, the increase of pro-apoptotic protein BIM expression in both mRNA and protein level has been illustrated. The recent findings provide an insight into the novel function of miR-101 in AML by activating BIM as an important mediator in intrinsic apoptosis pathways. Generally, miR-101 has been considered as a therapeutic target in our data and might have a valuable role in AML.
Collapse
Affiliation(s)
- Narges Nikoonahad Lotfabadi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.,Biology Department, Faculty of Sciences, Science and Arts University, Yazd, Iran
| | | | - Mohammad Hasan Sheikhha
- Reproductive & Genetic Unit, Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Biotechnology Research Center, International Campus, Shahid Sadoughi University of Medical Science, Yazd, Iran
| | - Seyed Mehdi Kalantar
- Reproductive & Genetic Unit, Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
8
|
Nebbioso A, Carafa V, Conte M, Tambaro FP, Abbondanza C, Martens J, Nees M, Benedetti R, Pallavicini I, Minucci S, Garcia-Manero G, Iovino F, Lania G, Ingenito C, Belsito Petrizzi V, Stunnenberg HG, Altucci L. c-Myc Modulation and Acetylation Is a Key HDAC Inhibitor Target in Cancer. Clin Cancer Res 2017; 23:2542-2555. [PMID: 27358484 DOI: 10.1158/1078-0432.ccr-15-2388] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 05/03/2016] [Accepted: 06/09/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Histone deacetylase inhibitors (HDACi) are promising anticancer drugs. Although some HDACi have entered the clinic, the mechanism(s) underlying their tumor selectivity are poorly understood.Experimental Design and Results: Using gene expression analysis, we define a core set of six genes commonly regulated in acute myeloid leukemia (AML) blasts and cell lines. MYC, the most prominently modulated, is preferentially altered in leukemia. Upon HDACi treatment, c-Myc is acetylated at lysine 323 and its expression decreases, leading to TRAIL activation and apoptosis. c-Myc binds to the TRAIL promoter on the proximal GC box through SP1 or MIZ1, impairing TRAIL activation. HDACi exposure triggers TRAIL expression, altering c-Myc-TRAIL binding. These events do not occur in normal cells. Excitingly, this inverse correlation between TRAIL and c-Myc is supported by HDACi treatment ex vivo of AML blasts and primary human breast cancer cells. The predictive value of c-Myc to HDACi responsiveness is confirmed in vivo in AML patients undergoing HDACi-based clinical trials.Conclusions: Collectively, our findings identify a key role for c-Myc in TRAIL deregulation and as a biomarker of the anticancer action of HDACi in AML. The potential improved patient stratification could pave the way toward personalized therapies. Clin Cancer Res; 23(10); 2542-55. ©2016 AACR.
Collapse
Affiliation(s)
- Angela Nebbioso
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy.
- Department of Molecular Biology, Faculties of Science and Medicine, Radboud University, Nijmegen Center for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Vincenzo Carafa
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy
| | | | - Francesco Paolo Tambaro
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy
- Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ciro Abbondanza
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy
| | - Joost Martens
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy
- Department of Molecular Biology, Faculties of Science and Medicine, Radboud University, Nijmegen Center for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Matthias Nees
- VTT Technical Research Centre of Finland, Espoo, Finland
| | - Rosaria Benedetti
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy
| | - Isabella Pallavicini
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Saverio Minucci
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | | | - Francesco Iovino
- Dipartimento Scienze Anestesiologiche, Chirurgiche e dell'Emergenza, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy
| | - Gabriella Lania
- Institute of Genetics and Biophysics (IGB) 'Adriano Buzzati Traverso', Naples, Italy
| | | | | | - Hendrik G Stunnenberg
- Department of Molecular Biology, Faculties of Science and Medicine, Radboud University, Nijmegen Center for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Lucia Altucci
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy.
- Institute of Genetics and Biophysics (IGB) 'Adriano Buzzati Traverso', Naples, Italy
| |
Collapse
|
9
|
Jez M, Ciesla M, Stepniewski J, Langrzyk A, Muchova L, Vitek L, Jozkowicz A, Dulak J. Valproic acid downregulates heme oxygenase-1 independently of Nrf2 by increasing ubiquitination and proteasomal degradation. Biochem Biophys Res Commun 2017; 485:160-166. [PMID: 28189672 DOI: 10.1016/j.bbrc.2017.02.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 02/07/2017] [Indexed: 01/01/2023]
Abstract
AIMS Heme oxygenase-1 (HO-1; HMOX1 in human, Hmox1 in mice) is an antioxidative enzyme affecting wide range of sub-cellular processes. It was shown to modulate tumor growth or vascular-related diseases, thus being putative molecular target for tailored therapies. Therefore it is of importance to elucidate novel compounds regulating HO-1 activity/expression and to delineate mechanisms of their action. In the present study we aimed to understand mode of action of valproic acid (VA), an antiepileptic drug, on HO-1 expression. RESULTS We demonstrated that HO-1 expression is decreased by VA at protein but not mRNA level in human alveolar rhabdomyosarcoma cell line CW9019. Nrf2 transcription factor, the activator of HO-1 expression through ARE sequence, was excluded as a mediator of HO-1 decrease, as VA downregulated Bach1, a Nrf2 repressor, concomitantly upregulating ARE activation. Also miRNA-dependent inhibition was excluded as a mechanism of HMOX1 regulation. However, co-immunoprecipitation assay showed a higher level of ubiquitinated HO-1 after VA treatment. Accordingly, MG132, an inhibitor of proteasomal degradation, reversed the effect of VA on HO-1 suggesting that decrease in HO-1 expression by VA is through protein stability. The inhibitory effect of VA on HO-1 was also observed in murine cells including embryonic fibroblasts isolated from Nrf2-deficient mice, what confirms Nrf2-independent effect of the compound. Importantly, VA decreased also HO-1 expression and activity in murine skeletal muscles in vivo. CONCLUSION Our data indicate that VA downregulates HO-1 by acting through ubiquitin-proteasomal pathway leading to decrease in protein level.
Collapse
Affiliation(s)
- Mateusz Jez
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maciej Ciesla
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jacek Stepniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | | | - Lucie Muchova
- Fourth Department of Internal Medicine and Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Libor Vitek
- Fourth Department of Internal Medicine and Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Kardio-Med Silesia, M. Curie-Skłodowskiej 10c, Zabrze, Poland.
| |
Collapse
|
10
|
Yu D, Yang X, Lu X, Shi L, Feng B. Ethyl acetate extract of Peperomia tetraphylla induces cytotoxicity, cell cycle arrest, and apoptosis in lymphoma U937 cells. Biomed Pharmacother 2016; 84:1802-1809. [PMID: 27847202 DOI: 10.1016/j.biopha.2016.10.092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 10/24/2016] [Accepted: 10/30/2016] [Indexed: 01/10/2023] Open
Abstract
The current study evaluated the cytotoxicity and the mechanism of apoptotic induction by Peperomia tetraphylla in U937 lymphoma cells. The results showed that P. tetraphylla ethyl acetate extract (EAEPT) inhibited the cell growth in U937 cells by MTT assay. After the U937 cells were treated with EAEPT, the cells exhibited marked morphological features of apoptosis (Hoechst 33342 staining) and the number of apoptotic cell (Annexin V-FITC/PI staining) increased. The treatment of EAEPT could induce loss of mitochondrial membrane potential (MMP) and increase the ROS level. Moreover, EAEPT treatment resulted in the accumulation of cells at S phase. We found that EAEPT could induce the cleavage of the caspase 3, caspase 8, caspase 9 and Bid. And the treatment of EAEPT could increase expression of Bax and down-regulate the expression of CCNB1, CCND1 and CDK1. The sub-fraction of EAEPT, namely EASub1 demonstrated the highest cytotoxicity activity on U937 cells. It was confirmed that EAEPT could inhibit the growth of U937 cells by blocking the cell cycle and prompted apoptosis via the ROS-medicated mitochondria pathway in vitro.
Collapse
Affiliation(s)
- Dayong Yu
- The School of Life Science and Biotechnology, Dalian University, Dalian 116622, PR China.
| | - Xiuxiu Yang
- The School of Life Science and Biotechnology, Dalian University, Dalian 116622, PR China
| | - Xuan Lu
- The School of Life Science and Biotechnology, Dalian University, Dalian 116622, PR China
| | - Liying Shi
- The School of Life Science and Biotechnology, Dalian University, Dalian 116622, PR China
| | - Baomin Feng
- The School of Life Science and Biotechnology, Dalian University, Dalian 116622, PR China.
| |
Collapse
|
11
|
Liu T, Guo Q, Guo H, Hou S, Li J, Wang H. Quantitative analysis of histone H3 and H4 post-translational modifications in doxorubicin-resistant leukemia cells. Biomed Chromatogr 2015; 30:638-44. [DOI: 10.1002/bmc.3608] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 06/26/2015] [Accepted: 08/24/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Tao Liu
- International Joint Cancer Institute; Second Military Medical University; Shanghai China
| | - Qingcheng Guo
- International Joint Cancer Institute; Second Military Medical University; Shanghai China
- State Key Laboratory of Antibody Medicine and Targeted Therapy; Shanghai Key Laboratory of Cell Engineering; Shanghai China
| | - Huaizu Guo
- International Joint Cancer Institute; Second Military Medical University; Shanghai China
- State Key Laboratory of Antibody Medicine and Targeted Therapy; Shanghai Key Laboratory of Cell Engineering; Shanghai China
| | - Sheng Hou
- International Joint Cancer Institute; Second Military Medical University; Shanghai China
- PLA General Hospital Cancer Center; PLA Postgraduate School of Medicine; Beijing China
| | - Jing Li
- International Joint Cancer Institute; Second Military Medical University; Shanghai China
| | - Hao Wang
- International Joint Cancer Institute; Second Military Medical University; Shanghai China
- PLA General Hospital Cancer Center; PLA Postgraduate School of Medicine; Beijing China
| |
Collapse
|
12
|
Borutinskaitė V, Navakauskienė R. The Histone Deacetylase Inhibitor BML-210 Influences Gene and Protein Expression in Human Promyelocytic Leukemia NB4 Cells via Epigenetic Reprogramming. Int J Mol Sci 2015; 16:18252-69. [PMID: 26287160 PMCID: PMC4581243 DOI: 10.3390/ijms160818252] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/22/2015] [Accepted: 07/23/2015] [Indexed: 12/31/2022] Open
Abstract
Today, cancer is understood as an epigenetic as well as genetic disease. The main epigenetic hallmarks of the cancer cell are DNA methylation and histone modifications. Proteins such as histone deacetylases (HDACs) that cause modifications of histones and other proteins can be targets for novel anticancer agents. Recently, interest in compounds that can inhibit HDACs increased, and now there are many HDACs inhibitors (HDACIs) available with different chemical structures, biological and biochemical properties; hopefully some of them will succeed, probably in combination with other agents, in cancer therapies. In our study we focused on the novel HDACI-BML-210. We found that BML-210 (N-phenyl-N'-(2-Aminophenyl)hexamethylenediamide) inhibits the growth of NB4 cells in dose- and time-dependent manner. In this study we also examined how expression and activity of HDACs are affected after leukemia cell treatment with BML-210. Using a mass spectrometry method we identified proteins that changed expression after treatment with BML-210. We prepared RT-PCR analysis of these genes and the results correlated with proteomic data. Based on these and other findings from our group, we suggest that HDACIs, like BML-210, can be promising anticancer agents in promyelocytic leukemia treatment.
Collapse
Affiliation(s)
- Veronika Borutinskaitė
- Department of Molecular Cell Biology, Institute of Biochemistry, Vilnius University, Mokslininkų 12, Vilnius LT 08662, Lithuania.
| | - Rūta Navakauskienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Vilnius University, Mokslininkų 12, Vilnius LT 08662, Lithuania.
| |
Collapse
|
13
|
Ilyas AM, Ahmad S, Faheem M, Naseer MI, Kumosani TA, Al-Qahtani MH, Gari M, Ahmed F. Next generation sequencing of acute myeloid leukemia: influencing prognosis. BMC Genomics 2015; 16 Suppl 1:S5. [PMID: 25924101 PMCID: PMC4315161 DOI: 10.1186/1471-2164-16-s1-s5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clonal disorder of the blood forming cells characterized by accumulation of immature blast cells in the bone marrow and peripheral blood. Being a heterogeneous disease, AML has been the subject of numerous studies that focus on unraveling the clinical, cellular and molecular variations with the aim to better understand and treat the disease. Cytogenetic-risk stratification of AML is well established and commonly used by clinicians in therapeutic management of cases with chromosomal abnormalities. Successive inclusion of novel molecular abnormalities has substantially modified the classification and understanding of AML in the past decade. With the advent of next generation sequencing (NGS) technologies the discovery of novel molecular abnormalities has accelerated. NGS has been successfully used in several studies and has provided an unprecedented overview of molecular aberrations as well as the underlying clonal evolution in AML. The extended spectrum of abnormalities discovered by NGS is currently under extensive validation for their prognostic and therapeutic values. In this review we highlight the recent advances in the understanding of AML in the NGS era.
Collapse
|
14
|
Sui X, Kong N, Wang Z, Pan H. Epigenetic regulation of the human telomerase reverse transciptase gene: A potential therapeutic target for the treatment of leukemia (Review). Oncol Lett 2013; 6:317-322. [PMID: 24137323 PMCID: PMC3789043 DOI: 10.3892/ol.2013.1367] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 05/17/2013] [Indexed: 12/21/2022] Open
Abstract
Telomerase activation is a critical step in human carcinogenesis through the maintenance of telomeres. Telomerase activity is primarily regulated by the human telomerase reverse transcriptase gene (hTERT), thus, an improved understanding of the transcriptional control of hTERT may provide potential therapeutic targets for the treatment of leukemia and other forms of cancer. Epigenetic modulation, a significant regulatory process in cell biology, has recently been shown to be involved in the regulation of the hTERT gene. Moreover, several epigenetic modifiers, including DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors, are now in pre- and early clinical trials of leukemia as monotherapies or in combination with other drugs, and have achieved significant clinical success. In the present review, the epigenetic mechanisms associated with telomerase activity in leukemia, and the therapeutic potential of an antitelomerase strategy that combines epigenetic modifiers with telomerase hTR subunit small molecule inhibitors are discussed.
Collapse
Affiliation(s)
- Xinbing Sui
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, P.R. China
| | | | | | | |
Collapse
|
15
|
Florean C, Schnekenburger M, Grandjenette C, Dicato M, Diederich M. Epigenomics of leukemia: from mechanisms to therapeutic applications. Epigenomics 2012; 3:581-609. [PMID: 22126248 DOI: 10.2217/epi.11.73] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Leukemogenesis is a multistep process in which successive transformational events enhance the ability of a clonal population arising from hematopoietic progenitor cells to proliferate, differentiate and survive. Clinically and pathologically, leukemia is subdivided into four main categories: chronic lymphocytic leukemia, chronic myeloid leukemia, acute lymphocytic leukemia and acute myeloid leukemia. Leukemia has been previously considered only as a genetic disease. However, in recent years, significant advances have been made in the elucidation of the leukemogenesis-associated processes. Thus, we have come to understand that epigenetic alterations including DNA methylation, histone modifications and miRNA are involved in the permanent changes of gene expression controlling the leukemia phenotype. In this article, we will focus on the epigenetic defects associated with leukemia and their implications as biomarkers for diagnostic, prognostic and therapeutic applications.
Collapse
Affiliation(s)
- Cristina Florean
- Laboratoire de Biologie Moléculaire et Cellulaire de Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | | | | | | | | |
Collapse
|
16
|
Dell'Aversana C, Lepore I, Altucci L. HDAC modulation and cell death in the clinic. Exp Cell Res 2012; 318:1229-44. [PMID: 22336671 DOI: 10.1016/j.yexcr.2012.01.025] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 01/26/2012] [Accepted: 01/27/2012] [Indexed: 01/29/2023]
Abstract
Histone acetyltransferases (HATs) and histone deacetylases (HDACs) are two opposing classes of enzymes, which finely regulate the balance of histone acetylation affecting chromatin packaging and gene expression. Imbalanced acetylation has been associated with carcinogenesis and cancer progression. In contrast to genetic mutations, epigenetic changes are potentially reversible. This implies that epigenetic alterations are amenable to pharmacological interventions. Accordingly, some epigenetic-based drugs (epidrugs) have been approved by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for cancer treatment. Here, we focus on the biological features of HDAC inhibitors (HDACis), analyzing the mechanism(s) of action and their current use in clinical practice.
Collapse
|
17
|
Chaib H, Nebbioso A, Prebet T, Castellano R, Garbit S, Restouin A, Vey N, Altucci L, Collette Y. Anti-leukemia activity of chaetocin via death receptor-dependent apoptosis and dual modulation of the histone methyl-transferase SUV39H1. Leukemia 2011; 26:662-74. [PMID: 21979880 DOI: 10.1038/leu.2011.271] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Epigenetic deregulation is involved in acute myeloid leukemia (AML) pathogenesis and epigenetic targeting drugs are in clinical trial. Since the first results with histone-deacetylase inhibitors in AML are controversial, novel single and combined treatments need to be explored. It is tempting to combine chromatin-targeting drugs. SUV39H1, the main methyl-transferase for lysine 9 tri-methylation on histone H3, interacts with oncogenes involved in AML and acts as a transcriptional repressor for hematopoietic differentiation and immortalization. We report here that pharmacological inhibition of SUV39H1 by chaetocin induces apoptosis in leukemia cell lines in vitro and primary AML cells ex vivo, and that it interferes with leukemia growth in vivo. Chaetocin treatment upregulates reactive oxygen species (ROS) production as well as the transcription of death-receptor-related genes, in a ROS-dependent manner, leading to death receptor-dependent apoptosis. In addition to its direct inhibition by chaetocin, SUV39H1 is indirectly modulated by chaetocin-induced ROS. Accordingly, chaetocin potentiates other anti-AML drugs, in a ROS-dependent manner. The decryption of a dual mechanism of action against AML involving both direct and indirect SUV39H1 modulation represents an innovative read-out for the anticancer activity of chaetocin and for its synergy with other anti-AML drugs, suggesting new therapeutic combination strategies in AML.
Collapse
Affiliation(s)
- H Chaib
- Inserm, U891, Centre de Recherche en Cancérologie de Marseille, Marseille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Souto JA, Vaz E, Lepore I, Pöppler AC, Franci G, Álvarez R, Altucci L, de Lera ÁR. Synthesis and Biological Characterization of the Histone Deacetylase Inhibitor Largazole and C7- Modified Analogues. J Med Chem 2010; 53:4654-67. [DOI: 10.1021/jm100244y] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- José A. Souto
- Departamento de Química Orgánica, Universidade de Vigo, Lagoas-Marcosende, 36310 Vigo, Spain
| | - Esther Vaz
- Departamento de Química Orgánica, Universidade de Vigo, Lagoas-Marcosende, 36310 Vigo, Spain
| | - Ilaria Lepore
- Dipartimento di Patologia Generale, Seconda Università degli Studi di Napoli, Vico L. de Crecchio 7, 80138 Napoli, Italy
| | - Ann-Christin Pöppler
- Departamento de Química Orgánica, Universidade de Vigo, Lagoas-Marcosende, 36310 Vigo, Spain
| | - Gianluigi Franci
- Dipartimento di Patologia Generale, Seconda Università degli Studi di Napoli, Vico L. de Crecchio 7, 80138 Napoli, Italy
- IGB-CNR, via Pietro Castellino, Napoli, Italy
| | - Rosana Álvarez
- Departamento de Química Orgánica, Universidade de Vigo, Lagoas-Marcosende, 36310 Vigo, Spain
| | - Lucia Altucci
- Dipartimento di Patologia Generale, Seconda Università degli Studi di Napoli, Vico L. de Crecchio 7, 80138 Napoli, Italy
- IGB-CNR, via Pietro Castellino, Napoli, Italy
| | - Ángel R. de Lera
- Departamento de Química Orgánica, Universidade de Vigo, Lagoas-Marcosende, 36310 Vigo, Spain
| |
Collapse
|
19
|
Borbone E, Berlingieri MT, De Bellis F, Nebbioso A, Chiappetta G, Mai A, Altucci L, Fusco A. Histone deacetylase inhibitors induce thyroid cancer-specific apoptosis through proteasome-dependent inhibition of TRAIL degradation. Oncogene 2009; 29:105-16. [DOI: 10.1038/onc.2009.306] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
20
|
Hoves S, Aigner M, Pfeiffer C, Laumer M, Obermann EC, Mackensen A. In situ analysis of the antigen-processing machinery in acute myeloid leukaemic blasts by tissue microarray. Leukemia 2009; 23:877-85. [DOI: 10.1038/leu.2008.391] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
21
|
Mai A, Valente S, Nebbioso A, Simeoni S, Ragno R, Massa S, Brosch G, De Bellis F, Manzo F, Altucci L. New pyrrole-based histone deacetylase inhibitors: binding mode, enzyme- and cell-based investigations. Int J Biochem Cell Biol 2008; 41:235-47. [PMID: 18834955 DOI: 10.1016/j.biocel.2008.09.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Revised: 09/03/2008] [Accepted: 09/03/2008] [Indexed: 11/30/2022]
Abstract
Aroyl-pyrrolyl-hydroxy-amides (APHAs) are a class of synthetic HDAC inhibitors described by us since 2001. Through structure-based drug design, two isomers of the APHA lead compound 1, the 3-(2-benzoyl-1-methyl-1H-pyrrol-4-yl)-N-hydroxy-2-propenamide 2 and the 3-(2-benzoyl-1-methyl-1H-pyrrol-5-yl)-N-hydroxy-2-propenamide 3 (iso-APHAs) were designed, synthesized and tested in murine leukemia cells as antiproliferative and cytodifferentiating agents. To improve their HDAC activity and selectivity, chemical modifications at the benzoyl moieties were investigated and evaluated using three maize histone deacetylases: HD2, HD1-B (class I human HDAC homologue), and HD1-A (class II human HDAC homologue). Docking experiments on HD1-A and HD1-B homology models revealed that the different compounds selectivity profiles could be addressed to different binding modes as observed for the reference compound SAHA. Smaller hydrophobic cap groups improved class II HDAC selectivity through the interaction with HD1-A Asn89-Ser90-Ile91, while bulkier aromatic substituents increased class I HDAC selectivity. Taking into account the whole enzyme data and the functional test results, the described iso-APHAs showed a behaviour of class I/IIb HDACi, with 4b and 4i preferentially inhibiting class IIb and class I HDACs, respectively. When tested in the human leukaemia U937 cell line, 4i showed altered cell cycle (S phase arrest), joined to high (51%) apoptosis induction and significant (21%) differentiation activity.
Collapse
Affiliation(s)
- Antonello Mai
- Dipartimento di Studi Farmaceutici, Sapienza Università di Roma, 00185 Roma, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Su H, Nebbioso A, Carafa V, Chen Y, Yang B, Altucci L, You Q. Design, synthesis and biological evaluation of novel compounds with conjugated structure as anti-tumor agents. Bioorg Med Chem 2008; 16:7992-8002. [DOI: 10.1016/j.bmc.2008.07.066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Revised: 07/22/2008] [Accepted: 07/23/2008] [Indexed: 10/21/2022]
|
23
|
Epi-drugs to fight cancer: from chemistry to cancer treatment, the road ahead. Int J Biochem Cell Biol 2008; 41:199-213. [PMID: 18790076 DOI: 10.1016/j.biocel.2008.08.020] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Revised: 08/08/2008] [Accepted: 08/08/2008] [Indexed: 12/16/2022]
Abstract
In addition to genetic events, a variety of epigenetic events have been widely reported to contribute to the onset of many diseases including cancer. DNA methylation and histone modifications (such as acetylation, methylation, sumoylation, and phosphorylation) involving chromatin remodelling are among the most studied epigenetic mechanisms for regulation of gene expression leading, when altered, to some diseases. Epigenetic therapy tries to reverse the aberrations followed to the disruption of the balance of the epigenetic signalling ways through the use of both natural compounds and synthetic molecules, active on specific epi-targets. Such epi-drugs are, for example, inhibitors of DNA methyltransferases, histone deacetylases, histone acetyltransferases, histone methyltransferases, and histone demethylases. In this review we will focus on the chemical aspects of such molecules, joined to their effective (or potential) application in cancer therapy.
Collapse
|
24
|
Nolan L, Johnson PWM, Ganesan A, Packham G, Crabb SJ. Will histone deacetylase inhibitors require combination with other agents to fulfil their therapeutic potential? Br J Cancer 2008; 99:689-94. [PMID: 18728657 PMCID: PMC2528143 DOI: 10.1038/sj.bjc.6604557] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Histone deacetylase inhibitors have progressed rapidly from the laboratory to clinical testing. This review highlights the promising data for their combination with a wide range of established and novel anticancer agents and discusses the mechanisms that underpin these interactions.
Collapse
Affiliation(s)
- L Nolan
- Cancer Research UK Clinical Centre, School of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | | | | | | | | |
Collapse
|
25
|
Drogaris P, Wurtele H, Masumoto H, Verreault A, Thibault P. Comprehensive Profiling of Histone Modifications Using a Label-Free Approach and Its Applications in Determining Structure−Function Relationships. Anal Chem 2008; 80:6698-707. [DOI: 10.1021/ac800739d] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Paul Drogaris
- Institut de Recherche en Immunologie et Cancer, Department of Chemistry, and Department of Pathology and Cell Biology, Université de Montréal, BP 6128, Station Centre-ville, Montréal, Canada, H3C 3J7, and Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Ten-noudai, Tsukuba, Ibaraki, Japan 305-8572
| | - Hugo Wurtele
- Institut de Recherche en Immunologie et Cancer, Department of Chemistry, and Department of Pathology and Cell Biology, Université de Montréal, BP 6128, Station Centre-ville, Montréal, Canada, H3C 3J7, and Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Ten-noudai, Tsukuba, Ibaraki, Japan 305-8572
| | - Hiroshi Masumoto
- Institut de Recherche en Immunologie et Cancer, Department of Chemistry, and Department of Pathology and Cell Biology, Université de Montréal, BP 6128, Station Centre-ville, Montréal, Canada, H3C 3J7, and Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Ten-noudai, Tsukuba, Ibaraki, Japan 305-8572
| | - Alain Verreault
- Institut de Recherche en Immunologie et Cancer, Department of Chemistry, and Department of Pathology and Cell Biology, Université de Montréal, BP 6128, Station Centre-ville, Montréal, Canada, H3C 3J7, and Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Ten-noudai, Tsukuba, Ibaraki, Japan 305-8572
| | - Pierre Thibault
- Institut de Recherche en Immunologie et Cancer, Department of Chemistry, and Department of Pathology and Cell Biology, Université de Montréal, BP 6128, Station Centre-ville, Montréal, Canada, H3C 3J7, and Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Ten-noudai, Tsukuba, Ibaraki, Japan 305-8572
| |
Collapse
|
26
|
Scognamiglio A, Nebbioso A, Manzo F, Valente S, Mai A, Altucci L. HDAC-class II specific inhibition involves HDAC proteasome-dependent degradation mediated by RANBP2. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:2030-8. [PMID: 18691615 DOI: 10.1016/j.bbamcr.2008.07.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Revised: 07/06/2008] [Accepted: 07/10/2008] [Indexed: 12/20/2022]
Abstract
Discovered for their ability to deacetylate histones and repress transcription, HDACs are a promising target for therapy of human diseases. The class II HDACs are mainly involved in developmental and differentiation processes, such as myogenesis. We report here that class I and class II HDAC inhibitors such as SAHA or the class II selective inhibitor MC1568 induce down-regulation of class II HDACs in human cells. In particular, both SAHA and MC1568 induce HDAC 4 down-regulation by increasing its specific sumoylation followed by activation of proteasomal pathways of degradation. Sumoylation that corresponds to HDAC 4 nuclear localization results in a transient increase of the HDAC 4 repressive action on target genes such as RARalpha and TNFalpha. The HDAC 4 degradation that follows to its sumoylation results in gene target activation. Silencing of the RANBP2 E3 ligase reverts HDAC 4 repression by blocking its own sumoylation. These findings identify a crosstalk occurring between acetylation, deacetylation and sumoylation pathways and suggest that class II specific HDAC inhibitors may affect different epigenetic pathways.
Collapse
Affiliation(s)
- Annamaria Scognamiglio
- Dipartimento di Patologia Generale, Seconda Università degli Studi di Napoli, Vico L. De Crecchio 7, 80138 Napoli, Italy
| | | | | | | | | | | |
Collapse
|
27
|
Fournel M, Bonfils C, Hou Y, Yan PT, Trachy-Bourget MC, Kalita A, Liu J, Lu AH, Zhou NZ, Robert MF, Gillespie J, Wang JJ, Ste-Croix H, Rahil J, Lefebvre S, Moradei O, Delorme D, Macleod AR, Besterman JM, Li Z. MGCD0103, a novel isotype-selective histone deacetylase inhibitor, has broad spectrum antitumor activity in vitro and in vivo. Mol Cancer Ther 2008; 7:759-68. [PMID: 18413790 DOI: 10.1158/1535-7163.mct-07-2026] [Citation(s) in RCA: 257] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nonselective inhibitors of human histone deacetylases (HDAC) are known to have antitumor activity in mice in vivo, and several of them are under clinical investigation. The first of these, Vorinostat (SAHA), has been approved for treatment of cutaneous T-cell lymphoma. Questions remain concerning which HDAC isotype(s) are the best to target for anticancer activity and whether increased efficacy and safety will result with an isotype-selective HDAC inhibitor. We have developed an isotype-selective HDAC inhibitor, MGCD0103, which potently targets human HDAC1 but also has inhibitory activity against HDAC2, HDAC3, and HDAC11 in vitro. In intact cells, MGCD0103 inhibited only a fraction of the total HDAC activity and showed long-lasting inhibitory activity even upon drug removal. MGCD0103 induced hyperacetylation of histones, selectively induced apoptosis, and caused cell cycle blockade in various human cancer cell lines in a dose-dependent manner. MGCD0103 exhibited potent and selective antiproliferative activities against a broad spectrum of human cancer cell lines in vitro, and HDAC inhibitory activity was required for these effects. In vivo, MGCD0103 significantly inhibited growth of human tumor xenografts in nude mice in a dose-dependent manner and the antitumor activity correlated with induction of histone acetylation in tumors. Our findings suggest that the isotype-selective HDAC inhibition by MGCD0103 is sufficient for antitumor activity in vivo and that further clinical investigation is warranted.
Collapse
Affiliation(s)
- Marielle Fournel
- Department of Molecular Biology, MethylGene, Inc., 7220 Frederick-Banting, Montreal, Quebec H4S 2A1, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Bhaskara S, Chyla BJ, Amann JM, Knutson SK, Cortez D, Sun ZW, Hiebert SW. Deletion of histone deacetylase 3 reveals critical roles in S phase progression and DNA damage control. Mol Cell 2008; 30:61-72. [PMID: 18406327 DOI: 10.1016/j.molcel.2008.02.030] [Citation(s) in RCA: 294] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Revised: 12/10/2007] [Accepted: 02/11/2008] [Indexed: 12/28/2022]
Abstract
Histone deacetylases (HDACs) are enzymes that modify key residues in histones to regulate chromatin architecture, and they play a vital role in cell survival, cell-cycle progression, and tumorigenesis. To understand the function of Hdac3, a critical component of the N-CoR/SMRT repression complex, a conditional allele of Hdac3 was engineered. Cre-recombinase-mediated inactivation of Hdac3 led to a delay in cell-cycle progression, cell-cycle-dependent DNA damage, and apoptosis in mouse embryonic fibroblasts (MEFs). While no overt defects in mitosis were observed in Hdac3-/- MEFs, including normal H3Ser10 phosphorylation, DNA damage was observed in Hdac3-/- interphase cells, which appears to be associated with defective DNA double-strand break repair. Moreover, we noted that Hdac3-/- MEFs were protected from DNA damage when quiescent, which may provide a mechanistic basis for the action of HDAC inhibitors on cycling tumor cells.
Collapse
Affiliation(s)
- Srividya Bhaskara
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Krämer OH, Müller S, Buchwald M, Reichardt S, Heinzel T. Mechanism for ubiquitylation of the leukemia fusion proteins AML1-ETO and PML-RARalpha. FASEB J 2007; 22:1369-79. [PMID: 18073335 DOI: 10.1096/fj.06-8050com] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The chromosomal translocation products AML1-ETO and PML-RARalpha contribute to the pathogenesis of leukemias. Here, we demonstrate that both AML1-ETO and PML-RARalpha are degraded by the ubiquitin-proteasome system and that their turnover critically depends on the E2-conjugase UbcH8 and the E3-ligase SIAH-1. Contrary to its role in HDAC2 degradation, the E3-ligase RLIM does not target AML1-ETO and PML-RARalpha for ubiquitin-dependent elimination. RLIM rather is a substrate of SIAH-1, which indicates that these E3-ligases operate in a hierarchical order. Remarkably, proteasomal degradation of leukemia fusion proteins, in addition to the block of histone deacetylase (HDAC) enzymatic activity is a consequence of HDAC-inhibitor treatment. The former requires the induction of UbcH8 expression and each of these processes might be beneficial for leukemia treatment. Our observations shed light on the mechanism determining the interplay between E2-conjugases, E3-ligases, and their substrates and suggest a strategy for utilizing the ubiquitylation machinery in a therapeutic setting.
Collapse
Affiliation(s)
- Oliver H Krämer
- Institute of Biochemistry and Biophysics, University of Jena, Philosophenweg 12, D-07743 Jena, Germany.
| | | | | | | | | |
Collapse
|
30
|
Altucci L, Leibowitz MD, Ogilvie KM, de Lera AR, Gronemeyer H. RAR and RXR modulation in cancer and metabolic disease. Nat Rev Drug Discov 2007; 6:793-810. [PMID: 17906642 DOI: 10.1038/nrd2397] [Citation(s) in RCA: 410] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Retinoic acid receptors (RARs) are ligand-controlled transcription factors that function as heterodimers with retinoid X receptors (RXRs) to regulate cell growth and survival. The success of RAR modulation in the treatment of acute promyelocytic leukaemia (APL) has stimulated considerable interest in the development of RAR and RXR modulators. This has been aided by recent advances in the understanding of the biological role of RARs and RXRs and in the design of selective receptor modulators that might overcome the limitations of current drugs. Here, we discuss the challenges and opportunities for therapeutic strategies based on RXR and RAR modulators, with a focus on cancer and metabolic diseases such as diabetes and obesity.
Collapse
Affiliation(s)
- Lucia Altucci
- Dipartimento di Patologia Generale, Seconda Università degli Studi di Napoli, Vico Luigi de Crecchio 7, 80138 Napoli, Italy
| | | | | | | | | |
Collapse
|
31
|
Lohrum M, Stunnenberg HG, Logie C. The new frontier in cancer research: Deciphering cancer epigenetics. Int J Biochem Cell Biol 2007; 39:1450-61. [PMID: 17442611 DOI: 10.1016/j.biocel.2007.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2007] [Revised: 03/12/2007] [Accepted: 03/13/2007] [Indexed: 12/13/2022]
Abstract
Cancer has long been known to be a disease caused by alterations in the genetic blueprint of cells. In the past decade it has become apparent that epigenetic alterations also underlie the etiology of cancer. Since epigenetic changes may be more facile to reverse than genetic lesions, much research has been invested in their characterization. Success has indeed been booked in the clinic with drugs that erase DNA methylation imprints or that target histone post-translational modifications such as lysine acetylation. However, the actual consequences of current epigenetic pharmacological intervention protocols are still poorly characterized and may be rather pleiotropic in nature. The challenge we face is therefore to define the cellular enzymes responsible for epigenetic modifications at given genes under specific conditions, so as to develop pharmacological agents that target tumorigenic epigenetic lesions while eliciting minimal unwanted side effects. Application of genome-wide analytical tools has begun to provide spatio-temporally resolved data that will be crucial to achieve this goal. Finally, the molecular mode of action of epigenetic drugs may be more intricate than initially thought, involving more than DNA and histones, since it has been reported that transcription (co)factors are themselves also targeted by histone modifying enzymes.
Collapse
Affiliation(s)
- Marion Lohrum
- Molecular Biology Department, Nijmegen Centre for Molecular Life Sciences, Radboud University, The Netherlands
| | | | | |
Collapse
|
32
|
Tsapis M, Lieb M, Manzo F, Shankaranarayanan P, Herbrecht R, Lutz P, Gronemeyer H. HDAC inhibitors induce apoptosis in glucocorticoid-resistant acute lymphatic leukemia cells despite a switch from the extrinsic to the intrinsic death pathway. Int J Biochem Cell Biol 2007; 39:1500-9. [PMID: 17499001 DOI: 10.1016/j.biocel.2007.03.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Revised: 03/09/2007] [Accepted: 03/09/2007] [Indexed: 10/23/2022]
Abstract
Inhibitors of histone deacetylases (HDACi's) are promising novel tools for cancer therapy. We have compared the growth inhibitory and apoptogenic potential of the pan-HDACi SAHA and the sub-class I selective HDAC inhibitor MS275, as well as valproic acid (VPA) on glucocorticoid sensitive and resistant B (B-ALL) and T (T-ALL) cell acute lymphoblastic leukemia cells and patients blasts. In contrast, to our previous results with U937 acute myeloid leukemia (AML) cells which showed a similar activity of MS275 and SAHA in growth inhibition and apoptosis induction, both B and T-ALL cells were much more efficiently killed by SAHA and VPA than by MS275. The same relative potency was observed with some patient ALL blasts treated ex vivo. SAHA displayed similar efficacy on glucocorticoid-sensitive and insensitive ALL cells but did not synergize with dexamethasone. In studying mediators of apoptosis we found that the TRAIL receptor DR5 is constitutively expressed in glucocorticoid-sensitive CEM-C7 cells which are also TRAIL sensitive. In contrast, glucocorticoid-insensitive CEM-C1 cells do not express DR5 and are insensitive to TRAIL. However, SAHA induces, in addition to p21(WAF1/CIP1) also re-expression of DR5. Importantly, SAHA-induced apoptosis of CEM-C7 cells operates through initiator caspase 10, while it induces apoptosis of CEM-C1 cells through the intrinsic, as well as through caspase-independent death pathways. Our data suggest that the generation of resistance to glucocorticoids has dramatically altered death signaling in these cells and that SAHA overcomes these restrictions by inducing alternative death pathways.
Collapse
Affiliation(s)
- Michael Tsapis
- Department of Cell Biology and Signal Transduction, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)/CNRS/INSERM/ULP, BP 10142, F-67404 Illkirch Cedex, C.U. de Strasbourg, France
| | | | | | | | | | | | | |
Collapse
|
33
|
Vitoux D, Nasr R, de The H. Acute promyelocytic leukemia: New issues on pathogenesis and treatment response. Int J Biochem Cell Biol 2007; 39:1063-70. [PMID: 17468032 DOI: 10.1016/j.biocel.2007.01.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Revised: 12/21/2006] [Accepted: 01/01/2007] [Indexed: 11/15/2022]
Abstract
Pathogenesis of acute promyelocytic leukemia appears to be one of the best understood among human malignancies. The ability of retinoic acid (RA) and arsenic trioxide to directly target the oncogenic promyelocytic leukemia-retinoic receptor A (PML-RARA) fusion protein also made this disease the first model for oncogene-targeted therapies. A set of recent data has significantly increased the complexity of our view of acute promyelocytic leukemia pathogenesis, as well as of therapeutic response. This review summarizes and discusses these findings, which yield novels questions and models.
Collapse
MESH Headings
- Arsenic Trioxide
- Arsenicals/pharmacology
- Arsenicals/therapeutic use
- Humans
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/pathology
- Models, Biological
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Oxides/pharmacology
- Oxides/therapeutic use
- Tretinoin/pharmacology
- Tretinoin/therapeutic use
Collapse
Affiliation(s)
- Dominique Vitoux
- CNRS UMR 7151, Université Paris 7, Equipe labellisée par la Ligue Nationale contre le Cancer, Hôpital Saint-Louis (APHP), 1 av Claude Vellefaux, 75475 Paris Cedex 10, France.
| | | | | |
Collapse
|
34
|
Germain P, Chambon P, Eichele G, Evans RM, Lazar MA, Leid M, De Lera AR, Lotan R, Mangelsdorf DJ, Gronemeyer H. International Union of Pharmacology. LX. Retinoic acid receptors. Pharmacol Rev 2006; 58:712-25. [PMID: 17132850 DOI: 10.1124/pr.58.4.4] [Citation(s) in RCA: 297] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Retinoid is a term for compounds that bind to and activate retinoic acid receptors (RARalpha, RARbeta, and RARgamma), members of the nuclear hormone receptor superfamily. The most important endogenous retinoid is all-trans-retinoic acid. Retinoids regulate a wide variety of essential biological processes, such as vertebrate embryonic morphogenesis and organogenesis, cell growth arrest, differentiation and apoptosis, and homeostasis, as well as their disorders. This review summarizes the considerable amount of knowledge generated on these receptors.
Collapse
Affiliation(s)
- Pierre Germain
- Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université Louis Pasteur, Illkirch, Communauté Urbaine de Strasbourg, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|