1
|
Ashraf R, Adel M, Serya RAT, Ibrahim E, Haffez H, Soror S, Abouzid KAM. Design and synthesis of novel Hydroxamate and non-Hydroxamate HDAC inhibitors based on Chromone and Quinazolone scaffolds. Bioorg Chem 2025; 161:108514. [PMID: 40319810 DOI: 10.1016/j.bioorg.2025.108514] [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: 01/17/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025]
Abstract
The development of selective histone deacetylase (HDAC) inhibitors represents an encouraging approach for cancer therapy. In this study, we report design, synthesis, and biological evaluation of hydroxamate, amidoxime, and carboxylic acid-based derivatives as novel HDAC inhibitors. The synthesized compounds were assessed for their inhibitory activity against multiple HDAC isoforms, particularly HDAC6, 7, and 8. Compounds 13, 16, 20, and 26 exhibited potent and selective inhibition of HDAC6. Compound 26 exhibited the most potent inhibitory activity against HDAC6, with an IC50 value of 70 nM. Additionally, compounds 17 and 23 demonstrated significant broad-spectrum antiproliferative activity across various cancer cell lines compared to other tested derivatives. Furthermore, compounds 17 and 23 showed promising total pan-HDAC inhibitory activity. Subsequent biological studies revealed that compounds 13, 16, 17, 20, 23, and 26 induced a combination of early and late apoptosis along with necrosis. In silico studies, including molecular docking and ADME predictions, were also conducted. Collectively, these findings highlight the potential of these compounds as promising candidates for the development of a novel class of selective HDAC6 inhibitors in the future.
Collapse
Affiliation(s)
- Rosaline Ashraf
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Mai Adel
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Rabah A T Serya
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Esraa Ibrahim
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, 11795 Cairo, Egypt; Center of Scientific Excellence "Helwan Structural Biology Research, (HSBR)", Helwan University, 11795 Cairo, Egypt
| | - Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, 11795 Cairo, Egypt; Center of Scientific Excellence "Helwan Structural Biology Research, (HSBR)", Helwan University, 11795 Cairo, Egypt
| | - Sameh Soror
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, 11795 Cairo, Egypt
| | - Khaled A M Abouzid
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt.
| |
Collapse
|
2
|
San José-Enériz E, Gimenez-Camino N, Rabal O, Garate L, Miranda E, Gómez-Echarte N, García F, Charalampopoulou S, Sáez E, Vilas-Zornoza A, San Martín-Uriz P, Valcárcel LV, Barrena N, Alignani D, Tamariz-Amador LE, Pérez-Ruiz A, Hilscher S, Schutkowski M, Alfonso-Pierola A, Martinez-Calle N, Larrayoz MJ, Paiva B, Calasanz MJ, Muñoz J, Isasa M, Martin-Subero JI, Pineda-Lucena A, Oyarzabal J, Agirre X, Prósper F. Epigenetic-based differentiation therapy for Acute Myeloid Leukemia. Nat Commun 2024; 15:5570. [PMID: 38956053 PMCID: PMC11219871 DOI: 10.1038/s41467-024-49784-y] [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: 02/02/2023] [Accepted: 06/18/2024] [Indexed: 07/04/2024] Open
Abstract
Despite the development of novel therapies for acute myeloid leukemia, outcomes remain poor for most patients, and therapeutic improvements are an urgent unmet need. Although treatment regimens promoting differentiation have succeeded in the treatment of acute promyelocytic leukemia, their role in other acute myeloid leukemia subtypes needs to be explored. Here we identify and characterize two lysine deacetylase inhibitors, CM-444 and CM-1758, exhibiting the capacity to promote myeloid differentiation in all acute myeloid leukemia subtypes at low non-cytotoxic doses, unlike other commercial histone deacetylase inhibitors. Analyzing the acetylome after CM-444 and CM-1758 treatment reveals modulation of non-histone proteins involved in the enhancer-promoter chromatin regulatory complex, including bromodomain proteins. This acetylation is essential for enhancing the expression of key transcription factors directly involved in the differentiation therapy induced by CM-444/CM-1758 in acute myeloid leukemia. In summary, these compounds may represent effective differentiation-based therapeutic agents across acute myeloid leukemia subtypes with a potential mechanism for the treatment of acute myeloid leukemia.
Collapse
Affiliation(s)
- Edurne San José-Enériz
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Naroa Gimenez-Camino
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Obdulia Rabal
- Small-Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Leire Garate
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Estibaliz Miranda
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Nahia Gómez-Echarte
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Fernando García
- ProteoRed-ISCIII, Unidad de Proteómica, Centro Nacional de Investigaciones Oncológicas (CNIO), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Stella Charalampopoulou
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Casanova 143, 08036, Barcelona, Spain
| | - Elena Sáez
- Small-Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Amaia Vilas-Zornoza
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Patxi San Martín-Uriz
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Luis V Valcárcel
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
- TECNUN, Universidad de Navarra, Manuel de Lardizábal 13, 20018, San Sebastián, Spain
| | - Naroa Barrena
- TECNUN, Universidad de Navarra, Manuel de Lardizábal 13, 20018, San Sebastián, Spain
| | - Diego Alignani
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Luis Esteban Tamariz-Amador
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
- Departmento de Hematología, Clínica Universidad de Navarra, and CCUN, Universidad de Navarra, Avenida Pío XII 36, 31008, Pamplona, Spain
| | - Ana Pérez-Ruiz
- Biomedical Engineering Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Sebastian Hilscher
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, 06120, Halle, Germany
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, 06120, Halle, Germany
| | - Mike Schutkowski
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, 06120, Halle, Germany
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, 06120, Halle, Germany
| | - Ana Alfonso-Pierola
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
- Departmento de Hematología, Clínica Universidad de Navarra, and CCUN, Universidad de Navarra, Avenida Pío XII 36, 31008, Pamplona, Spain
| | - Nicolás Martinez-Calle
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
- Departmento de Hematología, Clínica Universidad de Navarra, and CCUN, Universidad de Navarra, Avenida Pío XII 36, 31008, Pamplona, Spain
| | - María José Larrayoz
- CIMA LAB Diagnostics, Universidad de Navarra, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Bruno Paiva
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - María José Calasanz
- CIMA LAB Diagnostics, Universidad de Navarra, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Javier Muñoz
- Biocruces Bizkaia Health Research Institute, Cruces Plaza, 48903, Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009, Bilbao, Spain
| | - Marta Isasa
- ProteoRed-ISCIII, Unidad de Proteómica, Centro Nacional de Investigaciones Oncológicas (CNIO), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - José Ignacio Martin-Subero
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Casanova 143, 08036, Barcelona, Spain
- Departamento de Fundamentos Clínicos, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig de Lluís Companys 23, 08010, Barcelona, Spain
| | - Antonio Pineda-Lucena
- Small-Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Julen Oyarzabal
- Small-Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Avenida Pío XII 55, 31008, Pamplona, Spain.
| | - Xabier Agirre
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain.
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain.
| | - Felipe Prósper
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain.
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain.
- Departmento de Hematología, Clínica Universidad de Navarra, and CCUN, Universidad de Navarra, Avenida Pío XII 36, 31008, Pamplona, Spain.
| |
Collapse
|
3
|
Damiescu R, Efferth T, Dawood M. Dysregulation of different modes of programmed cell death by epigenetic modifications and their role in cancer. Cancer Lett 2024; 584:216623. [PMID: 38246223 DOI: 10.1016/j.canlet.2024.216623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/19/2023] [Accepted: 01/07/2024] [Indexed: 01/23/2024]
Abstract
Modifications of epigenetic factors affect our lives and can give important information regarding one's state of health. In cancer, epigenetic modifications play a crucial role, as they influence various programmed cell death types. The purpose of this review is to investigate how epigenetic modifications, such as DNA methylation, histone modifications, and non-coding RNAs, influence various cell death processes in suppressing or promoting cancer development. Autophagy and apoptosis are the most investigated programmed cell death modes, as based on the tumor stage these cell death types can either promote or prevent cancer evolution. Therefore, our discussion focuses on how epigenetic modifications affect autophagy and apoptosis, as well as their diagnostic and therapeutical potential in combination with available chemotherapeutics. Additionally, we summarize the available data regarding the role of epigenetic modifications on other programmed cell death modes, such as ferroptosis, necroptosis, and parthanatos in cancer and discuss current advancements.
Collapse
Affiliation(s)
- R Damiescu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, Mainz, Germany
| | - T Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, Mainz, Germany
| | - M Dawood
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, Mainz, Germany.
| |
Collapse
|
4
|
Zaalberg A, Pottendorfer E, Zwart W, Bergman AM. It Takes Two to Tango: The Interplay between Prostate Cancer and Its Microenvironment from an Epigenetic Perspective. Cancers (Basel) 2024; 16:294. [PMID: 38254784 PMCID: PMC10813511 DOI: 10.3390/cancers16020294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/28/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Prostate cancer is the second most common cancer in men worldwide and is associated with high morbidity and mortality. Consequently, there is an urgent unmet need for novel treatment avenues. In addition to somatic genetic alterations, deviations in the epigenetic landscape of cancer cells and their tumor microenvironment (TME) are critical drivers of prostate cancer initiation and progression. Unlike genomic mutations, epigenetic modifications are potentially reversible. Therefore, the inhibition of aberrant epigenetic modifications represents an attractive and exciting novel treatment strategy for castration-resistant prostate cancer patients. Moreover, drugs targeting the epigenome also exhibit synergistic interactions with conventional therapeutics by directly enhancing their anti-tumorigenic properties by "priming" the tumor and tumor microenvironment to increase drug sensitivity. This review summarizes the major epigenetic alterations in prostate cancer and its TME, and their involvement in prostate tumorigenesis, and discusses the impact of epigenome-targeted therapies.
Collapse
Affiliation(s)
- Anniek Zaalberg
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (A.Z.); (E.P.)
| | - Elisabeth Pottendorfer
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (A.Z.); (E.P.)
| | - Wilbert Zwart
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (A.Z.); (E.P.)
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Oncode Institute
| | - Andries M. Bergman
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (A.Z.); (E.P.)
- Division of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
5
|
Sengupta S, Ghufran SM, Khan A, Biswas S, Roychoudhury S. Transition of amyloid/mutant p53 from tumor suppressor to an oncogene and therapeutic approaches to ameliorate metastasis and cancer stemness. Cancer Cell Int 2022; 22:416. [PMID: 36567312 PMCID: PMC9791775 DOI: 10.1186/s12935-022-02831-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/11/2022] [Indexed: 12/27/2022] Open
Abstract
The tumor suppressor p53 when undergoes amyloid formation confers several gain-of-function (GOF) activities that affect molecular pathways crucial for tumorigenesis and progression like some of the p53 mutants. Even after successful cancer treatment, metastasis and recurrence can result in poor survival rates. The major cause of recurrence is mainly the remnant cancer cells with stem cell-like properties, which are resistant to any chemotherapy treatment. Several studies have demonstrated the role of p53 mutants in exacerbating cancer stemness properties and epithelial-mesenchymal transition in these remnant cancer cells. Analyzing the amyloid/mutant p53-mediated signaling pathways that trigger metastasis, relapse or chemoresistance may be helpful for the development of novel or improved individualized treatment plans. In this review, we discuss the changes in the metabolic pathways such as mevalonate pathway and different signaling pathways such as TGF-β, PI3K/AKT/mTOR, NF-κB and Wnt due to p53 amyloid formation, or mutation. In addition to this, we have discussed the role of the regulatory microRNAs and lncRNAs linked with the mutant or amyloid p53 in human malignancies. Such changes promote tumor spread, potential recurrence, and stemness. Importantly, this review discusses the cancer therapies that target either mutant or amyloid p53, restore wild-type functions, and exploit the synthetic lethal interactions with mutant p53.
Collapse
Affiliation(s)
- Shinjinee Sengupta
- grid.444644.20000 0004 1805 0217Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida, Uttar Pradesh, 201313 India
| | - Shaikh Maryam Ghufran
- grid.444644.20000 0004 1805 0217Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida, Uttar Pradesh, 201313 India
| | - Aqsa Khan
- grid.444644.20000 0004 1805 0217Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida, Uttar Pradesh, 201313 India
| | - Subhrajit Biswas
- grid.444644.20000 0004 1805 0217Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida, Uttar Pradesh, 201313 India
| | - Susanta Roychoudhury
- grid.489176.50000 0004 1803 6730Division of Research, Saroj Gupta Cancer Centre and Research Institute, Kolkata, 700063 India ,grid.417635.20000 0001 2216 5074Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
6
|
Sirtuin 4 Inhibits Prostate Cancer Progression and Metastasis by Modulating p21 Nuclear Translocation and Glutamate Dehydrogenase 1 ADP-Ribosylation. JOURNAL OF ONCOLOGY 2022; 2022:5498743. [PMID: 35847357 PMCID: PMC9283077 DOI: 10.1155/2022/5498743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/14/2022] [Accepted: 05/31/2022] [Indexed: 11/18/2022]
Abstract
Protein posttranslational modification regulates several biological mechanisms, including tumor progression. In this study, we show that the mitochondrial Sirtuin 4 (SIRT4), which has ADP-ribosylation activity, plays a role in prostate cancer (PCa) progression. Firstly, SIRT4 expression was verified in PCa tissues and cell lines by quantitative real-time PCR (qRT-PCR) and western blotting. Subsequently, we established stable PC-3 and 22rv1 cells that overexpressed SIRT4 and knocked down SIRT4, respectively. The functions of SIRT4 in PCa were explored through various phenotype experiments. The mechanism underlying the functions of SIRT4 was investigated through western blotting, immunoprecipitation, immunofluorescence, and nuclear and cytoplasmic extraction assays. We revealed that SIRT4 inhibited cell progression both in vivo and in vitro. Mechanistically, on the one hand, SIRT4 promoted the ADP-ribosylation of glutamate dehydrogenase 1 to inhibit the glutamine metabolism pathways. On the other hand, SIRT4 inhibited the phosphorylation of AKT, thereby affecting p21 phosphorylation and its cellular localization for cell cycle arrest. In conclusion, our study indicates that SIRT4 is directly associated with PCa progression and could be a novel target for PCa therapy.
Collapse
|
7
|
Shi Y, Xu S, Li S. Selinexor improves the anti-cancer effect of tucidinostat on TP53 wild-type breast cancer. Mol Cell Endocrinol 2022; 545:111558. [PMID: 35033575 DOI: 10.1016/j.mce.2022.111558] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/25/2021] [Accepted: 01/10/2022] [Indexed: 12/28/2022]
Abstract
Histone deacetylase (HDAC) is closely related to the initiation and development of breast cancer (BC). Its inhibitor (HDACi) has been used to treat BC, while the efficacy of clinical trials was not reached expectations. HDACi combined with other drugs may be an effective strategy. This study explored the effect of HDACi tucidinostat combined with selinexor, an exportin 1 (XPO1) inhibitor, on ER+Her2- BC cell lines of MCF-7 (wt-TP53), MDA-MB-175 (wt-TP53), MDA-MB-134 (mut-TP53) and T47D (mut-TP53) in vitro and cell derived xenografts (CDX) of MCF-7 in nude mice in vivo. Results showed that both tucidinostat and selinexor showed better inhibitory activities on wt-TP53 BC (MCF-7 and MDA-MB-175) comparing with mut-TP53 BC (MDA-MB-134 and T47D). Tucidinostat combined with selinexor significantly improved the effects of tucidinostat alone on the proliferation and invasion inhibitions and apoptosis promotions of MCF-7 and MDA-MB-175 cells in vitro. It also significantly enhanced the effects of tucidinostat on up-regulating the expression levels of acetyl-p53, nuclear p53, total p53, p21, Bax and Cleaved Caspase-3, and down-regulating the expression levels of Cyclin D1 and Bcl-2 in MCF-7 or MDA-MB-175 cells. Results consistent with in vitro were also obtained in CDX of MCF-7 in vivo. Taken together, we believe that tucidinostat and selinexor are potentially effective drug combinations for the treatment of wt-TP53 BC, and the molecular mechanism may be through enhancing the activity of p53 in the nucleus of BC cells to suppress proliferation and invasion and promote apoptosis.
Collapse
Affiliation(s)
- Yingfang Shi
- Breast Surgery, Jiujiang First People's Hospital, Jiujiang, Jiangxi Province, 332000, PR China
| | - Shengxi Xu
- Breast Surgery, Jiujiang First People's Hospital, Jiujiang, Jiangxi Province, 332000, PR China.
| | - Sen Li
- Breast Surgery, Jiujiang First People's Hospital, Jiujiang, Jiangxi Province, 332000, PR China
| |
Collapse
|
8
|
Aru B, Günay A, Demirel GY, Gürek AG, Atilla D. Evaluation of histone deacetylase inhibitor substituted zinc and indium phthalocyanines for chemo- and photodynamic therapy. RSC Adv 2021; 11:34963-34978. [PMID: 35494743 PMCID: PMC9042886 DOI: 10.1039/d1ra05404j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/14/2021] [Indexed: 01/03/2023] Open
Abstract
In this study, we synthesized and characterized 3-hydroxypyridin-2-thione (3-HPT) bearing zinc (ZnPc-1 and ZnPc-2) and indium (InPc-1 and InPc-2) phthalocyanine (Pc) derivatives, either non-peripherally or peripherally substituted as photosensitizer (PS) agents and evaluated their anti-cancer efficacy on two breast cancer cell lines, MDA-MB-231 and MCF-7 as well as a human endothelial cell line, HUVEC. Our results indicated different localization patterns between ZnPcs and InPcs in addition to enhanced effects on the mitochondrial network for InPcs. Moreover, peripheral or non-peripheral substitution of HDACi moieties altered cellular localization between ZnPc-1 and ZnPc-2, leading to increased IC50 values along with decreased anti-cancer activity for non-peripheral substitution. When considering the compounds' differential effects in vitro, our data indicates that further research is required to determine the ideal Pcs for anti-cancer PDT treatments since the core metals of the compounds have affected the cellular localization, and positioning of the chemotherapeutic residues may inhibit cellular penetrance.
Collapse
Affiliation(s)
- Başak Aru
- Faculty of Medicine, Immunology Department, Yeditepe University 34755 Ataşehir İstanbul Turkey
| | - Aysel Günay
- Department of Chemistry, Gebze Technical University 41400 Gebze Kocaeli Turkey
| | | | - Ayşe Gül Gürek
- Department of Chemistry, Gebze Technical University 41400 Gebze Kocaeli Turkey
| | - Devrim Atilla
- Department of Chemistry, Gebze Technical University 41400 Gebze Kocaeli Turkey
| |
Collapse
|
9
|
Li P, Cao W, Ding R, Cheng M, Xu X, Chen S, Chen B, Cao G, Xiong M. Expression and Prognostic Significance of Metastasis-Associated Protein 1 in Gastrointestinal Cancer. Front Oncol 2020; 10:542330. [PMID: 33409150 PMCID: PMC7780747 DOI: 10.3389/fonc.2020.542330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/09/2020] [Indexed: 01/30/2023] Open
Abstract
Background Metastasis-associated protein 1 (MTA1) has been considered as a transcriptional regulator, which is significantly related to the prognosis in various types of tumors. However, whether MTA1 is a potential prognostic index of gastrointestinal cancer (GIC) remains controversial. The current meta-analysis was performed to evaluate the role of MTA1 expression in the prediction of the clinicopathological features and survival in GIC cases. And the results of gastric cancer were verified by immunohistochemistry (IHC). Methods Eligible studies assessing the relationship between MTA1 and GIC by IHC were searched in the PubMed, Cochrane, Ovid, Web of Science and CNKI databases by various search strategies. The STATA 16.0 software was applied to gather data and to analyze the potential relationship between MTA1 and GIC. The expression level of MTA1 was examined in 80 GC samples by IHC assay. SPSS 20.0 was applied for statistical analysis, and the survival curves were calculated by the Kaplan-Meier method. The data of 95% CI was displayed as “[a-b]”. Results According to the meta-analysis, the expression level of MTA1 was tightly associated with the tumor size (OR=1.82 [1.16–2.84], P=0.009), tumor tissue differentiation (OR=1.71 [1.24–2.37], P=0.001), depth of invasion (OR=3.12 [2.55–3.83], P<0.001), lymphatic metastasis (OR=2.99 [2.02–4.43], P<0.001), distant metastasis (OR=4.66 [1.13–19.24], P=0.034), TNM stage (OR=4.28 [2.76–6.63], P<0.001). In addition, MTA1 played the negative effects in 1- (RR=2.48 [1.45–4.25], P=0.001), 3- (RR=1.66 [1.30–2.11], P<0.001) and 5-year (RR=1.73 [1.37–2.20], P<0.001). Study in subgroup, grouped by language and tumor type, we reached similar conclusions. Further validation by IHC yielded similar conclusions. Tumor size (P=0.008), lymph node metastasis (P=0.007) and distant metastasis (P=0.023) significantly accompanied with higher expression of MAT1 in GC cases. Besides, the expression level of MTA1 was statistically significantly correlated with OS in GC cases (HR=2.061 [1.066–3.986], P=0.032), which suggested that MTA1 might be an independent prognostic marker for GC. Finally, we verified the correlation between the expression level of MTA1 and prognosis of GC in 80 GC samples. Conclusions MTA1 is tightly associated with metastasis-related factors and may constitute a promising prognostic factor of GIC.
Collapse
Affiliation(s)
- Pengping Li
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of General Surgery, First People's Hospital of Xiaoshan, Hangzhou, China
| | - Wei Cao
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Rui Ding
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Emergency, the Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, China
| | - Mengqiu Cheng
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xin Xu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Sihan Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bo Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guodong Cao
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Maoming Xiong
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
10
|
The Role of Histone Acetylation-/Methylation-Mediated Apoptotic Gene Regulation in Hepatocellular Carcinoma. Int J Mol Sci 2020; 21:ijms21238894. [PMID: 33255318 PMCID: PMC7727670 DOI: 10.3390/ijms21238894] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/31/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023] Open
Abstract
Epigenetics, an inheritable phenomenon, which influences the expression of gene without altering the DNA sequence, offers a new perspective on the pathogenesis of hepatocellular carcinoma (HCC). Nonalcoholic steatohepatitis (NASH) is projected to account for a significant share of HCC incidence due to the growing prevalence of various metabolic disorders. One of the major molecular mechanisms involved in epigenetic regulation, post-translational histone modification seems to coordinate various aspects of NASH which will further progress to HCC. Mounting evidence suggests that the orchestrated events of cellular and nuclear changes during apoptosis can be regulated by histone modifications. This review focuses on the current advances in the study of acetylation-/methylation-mediated histone modification in apoptosis and the implication of these epigenetic regulations in HCC. The reversibility of epigenetic alterations and the agents that can target these alterations offers novel therapeutic approaches and strategies for drug development. Further molecular mechanistic studies are required to enhance information governing these epigenetic modulators, which will facilitate the design of more effective diagnosis and treatment options.
Collapse
|
11
|
Green SD, Konig H. Treatment of Acute Myeloid Leukemia in the Era of Genomics-Achievements and Persisting Challenges. Front Genet 2020; 11:480. [PMID: 32536937 PMCID: PMC7267060 DOI: 10.3389/fgene.2020.00480] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/17/2020] [Indexed: 01/15/2023] Open
Abstract
Acute myeloid leukemia (AML) represents a malignant disorder of the hematopoietic system that is mainly characterized by rapid proliferation, dysregulated apoptosis, and impaired differentiation of leukemic blasts. For several decades, the diagnostic approach in AML was largely based on histologic characteristics with little impact on the treatment decision-making process. This perspective has drastically changed within the past years due to the advent of novel molecular technologies, such as whole genome next-generation sequencing (NGS), and the resulting knowledge gain in AML biology and pathogenesis. After more than four decades of intensive chemotherapy as a "one-size-fits-all" concept, several targeted agents have recently been approved for the treatment of AML, either as single agents or as part of combined treatment regimens. Several other compounds, directed against regulators of apoptotic, epigenetic, or microenvironmental pathways, as well as modulators of the immune system, are currently in development and being investigated in clinical trials. The constant progress in AML research has started to produce improved survival rates and fueled hopes that a once rapidly fatal disease can be transformed into a chronic condition. In this review, the authors provide a summary of recent advances in the development of targeted AML therapies and discuss persistent challenges.
Collapse
Affiliation(s)
| | - Heiko Konig
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
12
|
Shankar E, Pandey M, Verma S, Abbas A, Candamo M, Kanwal R, Shukla S, MacLennan GT, Gupta S. Role of class I histone deacetylases in the regulation of maspin expression in prostate cancer. Mol Carcinog 2020; 59:955-966. [PMID: 32391971 DOI: 10.1002/mc.23214] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022]
Abstract
Maspin repression is frequently observed in prostate cancer; however, the molecular mechanism(s) causing the loss is not completely understood. Here, we demonstrate that inhibition of class I histone deacetylases (HDACs) mediates re-expression of maspin which plays an essential role in suppressing proliferation and migration capability in prostate cancer cells. Human prostate cancer LNCaP and DU145 cells treated with HDAC inhibitors, sodium butyrate, and trichostatin A, resulted in maspin re-expression. Interestingly, an exploration into the molecular mechanisms demonstrates that maspin repression in prostate tumor and human prostate cancer cell lines occurs via epigenetic silencing through an increase in HDAC activity/expression, independent of promoter DNA hypermethylation. Furthermore, transcriptional activation of maspin was accompanied with the suppression of HDAC1 and HDAC8 with significant p53 enrichment at the maspin promoter associated with an increase in histone H3/H4 acetylation. Our results provide evidence of maspin induction as a critical epigenetic event altered by class I HDACs in the restoration of balance to delay proliferation and migration ability of prostate cancer cells.
Collapse
Affiliation(s)
- Eswar Shankar
- Department of Urology, Case Western Reserve University, Cleveland, Ohio.,The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Mitali Pandey
- Department of Urology, Case Western Reserve University, Cleveland, Ohio.,The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Shiv Verma
- Department of Urology, Case Western Reserve University, Cleveland, Ohio.,The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Ata Abbas
- Department of Urology, Case Western Reserve University, Cleveland, Ohio.,The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Mario Candamo
- College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Rajnee Kanwal
- Department of Urology, Case Western Reserve University, Cleveland, Ohio.,The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Sanjeev Shukla
- Department of Urology, Case Western Reserve University, Cleveland, Ohio.,The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, Cleveland, Ohio.,The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio.,Department of Nutrition, Case Western Reserve University, Cleveland, Ohio.,Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, Ohio.,Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio
| |
Collapse
|
13
|
Lu T, Wei D, Yu K, Ma D, Xiong J, Fang Q, Wang J. Betulinic acid restores imatinib sensitivity in BCR-ABL1 kinase-independent, imatinib-resistant chronic myeloid leukemia by increasing HDAC3 ubiquitination and degradation. Ann N Y Acad Sci 2020; 1467:77-93. [PMID: 31930541 DOI: 10.1111/nyas.14298] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/28/2019] [Accepted: 12/18/2019] [Indexed: 12/14/2022]
Abstract
Although imatinib (IM) has been demonstrated to be an efficient treatment in chronic myeloid leukemia (CML), some patients still experience IM resistance and disease relapse. Through in vitro studies, we observed that HDAC3 levels were elevated in BCR-ABL1 kinase-independent, IM-resistant primary cells from CML patients and in IM-resistant K562 (K562R) cells and that downregulation of HDAC3 could enhance IM efficacy in K562R cells. Furthermore, betulinic acid (BA), a lupane-type pentacyclic triterpenoid saponin isolated from birch trees, restored IM sensitivity in the BCR-ABL1 kinase-independent, IM-resistant primary cells and in K562R cells, as well as in primary CD34+ bone marrow cells from CML patients. We found that BA restored IM sensitivity through inhibition of HDAC3 accumulation in cells, and that this was mediated by BA-dependent ubiquitination and degradation of HDAC3. BA at low dosage significantly increased IM antitumor effects on murine xenografts bearing K562R cells and inhibited HDAC3 expression in tumor tissue. Our findings demonstrated that HDAC3 is an essential factor in BCR-ABL1 kinase-independent IM resistance, and that BA in combination with IM may be a novel treatment strategy for overcoming IM resistance in CML.
Collapse
Affiliation(s)
- Tingting Lu
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Department of Clinical Laboratory Centre, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,School of Basic Medical Sciences, Guizhou Medical University, Guizhou, China
| | - Danna Wei
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| | - Kunlin Yu
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China.,Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Dan Ma
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| | - Jie Xiong
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| | - Qin Fang
- Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Jishi Wang
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| |
Collapse
|
14
|
Yerinde C, Siegmund B, Glauben R, Weidinger C. Metabolic Control of Epigenetics and Its Role in CD8 + T Cell Differentiation and Function. Front Immunol 2019; 10:2718. [PMID: 31849941 PMCID: PMC6901948 DOI: 10.3389/fimmu.2019.02718] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022] Open
Abstract
Epigenetic programs that control posttranslational modifications of histone proteins and DNA itself tightly regulate transcriptional networks determining the identity and function of CD8+ T cells. Chromatin-modifying enzymes such as histone acetyltransferases and deacetylases, represent key molecular determinants of the epigenetic imprinting of CD8+ T cells. The functions of these enzymes highly depend on the availability of key products of cellular metabolism pathways such as acetyl-CoA, NAD (Nicotinamide adenine dinucleotide) and SEM (S-adenosylmethionine), suggesting that there is a close crosstalk between the metabolic and the epigenetic regulation of CD8+ T cells. In this review, we will discuss the metabolic regulation of CD8+ T cell epigenetics during activation and differentiation. We will furthermore summarize how metabolic signals from the tumor microenvironment (TME) shape the epigenetic landscape of CD8+ T cells to better understand the mechanism underlying CD8+ T cell exhaustion in anti-tumor and anti-viral immunity, which might help to overcome limitations of current CD8+ T cell-based therapies.
Collapse
Affiliation(s)
- Cansu Yerinde
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Britta Siegmund
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Rainer Glauben
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Carl Weidinger
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Clinician Scientist Program, Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
15
|
San José-Enériz E, Gimenez-Camino N, Agirre X, Prosper F. HDAC Inhibitors in Acute Myeloid Leukemia. Cancers (Basel) 2019; 11:cancers11111794. [PMID: 31739588 PMCID: PMC6896008 DOI: 10.3390/cancers11111794] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/05/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by uncontrolled proliferation, differentiation arrest, and accumulation of immature myeloid progenitors. Although clinical advances in AML have been made, especially in young patients, long-term disease-free survival remains poor, making this disease an unmet therapeutic challenge. Epigenetic alterations and mutations in epigenetic regulators contribute to the pathogenesis of AML, supporting the rationale for the use of epigenetic drugs in patients with AML. While hypomethylating agents have already been approved in AML, the use of other epigenetic inhibitors, such as histone deacetylases (HDAC) inhibitors (HDACi), is under clinical development. HDACi such as Panobinostat, Vorinostat, and Tricostatin A have been shown to promote cell death, autophagy, apoptosis, or growth arrest in preclinical AML models, yet these inhibitors do not seem to be effective as monotherapies, but rather in combination with other drugs. In this review, we discuss the rationale for the use of different HDACi in patients with AML, the results of preclinical studies, and the results obtained in clinical trials. Although so far the results with HDACi in clinical trials in AML have been modest, there are some encouraging data from treatment with the HDACi Pracinostat in combination with DNA demethylating agents.
Collapse
Affiliation(s)
- Edurne San José-Enériz
- Área de Hemato-Oncología, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra (IDISNA), Universidad de Navarra, 31008 Pamplona, Spain; (E.S.J.-E.); (N.G.-C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Naroa Gimenez-Camino
- Área de Hemato-Oncología, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra (IDISNA), Universidad de Navarra, 31008 Pamplona, Spain; (E.S.J.-E.); (N.G.-C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Xabier Agirre
- Área de Hemato-Oncología, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra (IDISNA), Universidad de Navarra, 31008 Pamplona, Spain; (E.S.J.-E.); (N.G.-C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Correspondence: (X.A.); (F.P.); Tel.: +34-948-194700 (ext. 1002) (X.A.); +34-948-255400 (ext. 5807) (F.P.)
| | - Felipe Prosper
- Área de Hemato-Oncología, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra (IDISNA), Universidad de Navarra, 31008 Pamplona, Spain; (E.S.J.-E.); (N.G.-C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Departamento de Hematología, Clínica Universidad de Navarra, Universidad de Navarra, 31008 Pamplona, Spain
- Correspondence: (X.A.); (F.P.); Tel.: +34-948-194700 (ext. 1002) (X.A.); +34-948-255400 (ext. 5807) (F.P.)
| |
Collapse
|
16
|
Corton JC, Witt KL, Yauk CL. Identification of p53 Activators in a Human Microarray Compendium. Chem Res Toxicol 2019; 32:1748-1759. [PMID: 31397557 DOI: 10.1021/acs.chemrestox.9b00052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Biomarkers predictive of molecular and toxicological effects are needed to interpret emerging high-throughput transcriptomic data streams. The previously characterized 63 gene TGx-DDI biomarker that includes 20 genes known to be regulated by p53 was previously shown to accurately predict DNA damage in chemically treated cells. We comprehensively evaluated whether the molecular basis of the DDI predictions was based on a p53-dependent response. The biomarker was compared to microarray data in a compendium derived from human cells using the Running Fisher test, a nonparametric correlation test. Using the biomarker, we identified conditions that led to p53 activation, including exposure to the chemical nutlin-3 which disrupts interactions between p53 and the negative regulator MDM2 or by knockdown of MDM2. The expression of most of the genes in the biomarker (75%) were found to depend on p53 activation status based on gene behavior after TP53 overexpression or knockdown. The biomarker identified DDI chemicals that were strong inducers of p53 in wild-type cells; these p53 responses were decreased or abolished in cells after p53 knockdown by siRNAs. Using the biomarker, we screened ∼1950 chemicals in ∼9800 human cell line chemical vs control comparisons and identified ∼100 chemicals that caused p53 activation. Among the positive chemicals were many that are known to activate p53 through direct and indirect DNA damaging mechanisms. These results contribute to the evidence that the TGx-DDI biomarker is useful for identifying chemicals that cause DDI and activate p53.
Collapse
Affiliation(s)
- J Christopher Corton
- Integrated Systems Toxicology Division, NHEERL , United States Environmental Protection Agency , Research Triangle Park, Durham , North Carolina 27711 , United States
| | - Kristine L Witt
- Division of the National Toxicology Program , National Institute of Environmental Health Sciences , Research Triangle Park, Durham , North Carolina 27709 , United States
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Health Canada , Ottawa , Ontario K1A 0K9 , Canada
| |
Collapse
|
17
|
p53 at the Crossroads between Different Types of HDAC Inhibitor-Mediated Cancer Cell Death. Int J Mol Sci 2019; 20:ijms20102415. [PMID: 31096697 PMCID: PMC6567317 DOI: 10.3390/ijms20102415] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/06/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022] Open
Abstract
Cancer is a complex genetic and epigenetic-based disease that has developed an armada of mechanisms to escape cell death. The deregulation of apoptosis and autophagy, which are basic processes essential for normal cellular activity, are commonly encountered during the development of human tumors. In order to assist the cancer cell in defeating the imbalance between cell growth and cell death, histone deacetylase inhibitors (HDACi) have been employed to reverse epigenetically deregulated gene expression caused by aberrant post-translational protein modifications. These interfere with histone acetyltransferase- and deacetylase-mediated acetylation of both histone and non-histone proteins, and thereby exert a wide array of HDACi-stimulated cytotoxic effects. Key determinants of HDACi lethality that interfere with cellular growth in a multitude of tumor cells are apoptosis and autophagy, which are either mutually exclusive or activated in combination. Here, we compile known molecular signals and pathways involved in the HDACi-triggered induction of apoptosis and autophagy. Currently, the factors that determine the mode of HDACi-elicited cell death are mostly unclear. Correspondingly, we also summarized as yet established intertwined mechanisms, in particular with respect to the oncogenic tumor suppressor protein p53, that drive the interplay between apoptosis and autophagy in response to HDACi. In this context, we also note the significance to determine the presence of functional p53 protein levels in the cancer cell. The confirmation of the context-dependent function of autophagy will pave the way to improve the benefit from HDACi-mediated cancer treatment.
Collapse
|
18
|
Alp E, Damkaci F, Guven E, Tenniswood M. Starch nanoparticles for delivery of the histone deacetylase inhibitor CG-1521 in breast cancer treatment. Int J Nanomedicine 2019; 14:1335-1346. [PMID: 30863064 PMCID: PMC6388755 DOI: 10.2147/ijn.s191837] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background The efficacy of epigenetic drugs, such as histone deacetylase inhibitors, is often diminished by poor aqueous solubility resulting in limited bioavailability and a low therapeutic index. To overcome the suboptimal therapeutic index, we have developed a biocompatible starch nanoparticle formulation of CG-1521, a histone deacetylase inhibitor in preclinical development for hard-to-treat breast cancers, which improves its bioavailability and half-life. Methods The physicochemical parameters (size, zeta potential, morphology, loading, and release kinetics) of these nanoparticles (CG-NPs) have been optimized and their cytotoxic and apoptotic capacities measured in MCF-7 breast cancer cell line. The mechanism of action of the encapsulated drug was compared with the free drug at molecular level. Results We show that encapsulation of CG-1521 substantially reduces the release rate of drug and provides a significantly enhanced cytotoxic ability of nanoparticles compared with equivalent dose of free CG-1521. CG-NPs induced cell cycle arrest and significant apoptosis in MCF-7 cells in vitro. The biological action of encapsulated drug has the similar impact with free drug on gene expression. Conclusion The findings suggest that encapsulation of CG-1521 into starch nanoparticles can improve drug delivery of histone deacetylase inhibitors for breast cancer therapy without interfering with the mechanism of action of the drug.
Collapse
Affiliation(s)
- Esma Alp
- Department of Nanotechnology and Nanomedicine, Hacettepe University, Beytepe, Ankara 06800, Turkey.,Department of Chemistry, State University of New York at Oswego, Oswego, NY 13126, USA.,Cancer Research Center, Rensselaer, NY 12144, USA, .,Department of Biomedical Sciences, State University of New York, University at Albany, Rensselaer, NY 12144, USA,
| | - Fehmi Damkaci
- Department of Chemistry, State University of New York at Oswego, Oswego, NY 13126, USA
| | - Eylem Guven
- Department of Nanotechnology and Nanomedicine, Hacettepe University, Beytepe, Ankara 06800, Turkey
| | - Martin Tenniswood
- Cancer Research Center, Rensselaer, NY 12144, USA, .,Department of Biomedical Sciences, State University of New York, University at Albany, Rensselaer, NY 12144, USA,
| |
Collapse
|
19
|
Yuan Y, Hu Z, Bao M, Sun R, Long X, Long L, Li J, Wu C, Bao J. Screening of novel histone deacetylase 7 inhibitors through molecular docking followed by a combination of molecular dynamics simulations and ligand-based approach. J Biomol Struct Dyn 2018; 37:4092-4103. [PMID: 30417746 DOI: 10.1080/07391102.2018.1541141] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Histone acetylation/deacetylation is a key mechanism for transcription regulation which plays an important role in control of gene expression, tissue growth, and development. In particular, histone deacetylase 7 (HDAC7), a member of class IIa HDACs, is crucial to maintain cell homeostasis, and HDAC7 has emerged as a new target for cancer therapy. In this study, molecular docking was applied to screen candidate inhibitors and 21 compounds were found. Following the 50 ns molecular dynamics simulations and binding free energy calculation, ZINC00156160, ZINC01703144, ZINC04293665, and ZINC13900201 were identified as potential HDAC7 inhibitors, which would provide a sound starting point for further studies involving molecular modeling coupled with biochemical experiments. Meanwhile, similarity computation and substructure search were combined, and then we found that compounds sharing common backbone "CC(=O)N[C@@H](CSc1ccccc1)C(=O)O" could be efficient to inhibit the bioactivity of HDAC7. Then comparative molecular similarity indices analysis (CoMSIA) techniques were implemented to investigate the relationship between properties of the substituent group and bioactivities of small molecules. The CoMSIA model exhibited powerful predictivity, with satisfactory statistical parameters such as q2 of 0.659, R2 of 0.952, and F of 268.448. Contour maps of the CoMSIA model gave insight into the feature requirements of the common backbone for the HDAC7 inhibitory activity. Finally, details of designing novel HDAC7 inhibitors were confirmed by a combination of receptor-based docking and ligand-based structure-activity relationship. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yuan Yuan
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Zongyue Hu
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Minyue Bao
- b State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University , Chengdu , Sichuan , China
| | - Rong Sun
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Xin Long
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Li Long
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Jianzong Li
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Chuanfang Wu
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Jinku Bao
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China.,b State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University , Chengdu , Sichuan , China
| |
Collapse
|
20
|
A novel all-trans retinoic acid derivative inhibits proliferation and induces apoptosis of myelodysplastic syndromes cell line SKM-1 cells via up-regulating p53. Int Immunopharmacol 2018; 65:561-570. [DOI: 10.1016/j.intimp.2018.10.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/19/2018] [Accepted: 10/30/2018] [Indexed: 12/31/2022]
|
21
|
Andrade SN, Evangelista FCG, Seckler D, Marques DR, Freitas TR, Nunes RR, Oliveira JT, Ribeiro RIMA, Santos HB, Thomé RG, Taranto AG, Santos FV, Viana GHR, Freitas RP, Humberto JL, Sabino ADP, Hilário FF, Varotti FP. Synthesis, cytotoxic activity, and mode of action of new Santacruzamate A analogs. Med Chem Res 2018. [DOI: 10.1007/s00044-018-2244-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
22
|
Fuenmayor J, Cervera L, Rigau C, Gòdia F. Enhancement of HIV-1 VLP production using gene inhibition strategies. Appl Microbiol Biotechnol 2018; 102:4477-4487. [DOI: 10.1007/s00253-018-8930-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/06/2018] [Accepted: 03/10/2018] [Indexed: 10/17/2022]
|
23
|
ID4 regulates transcriptional activity of wild type and mutant p53 via K373 acetylation. Oncotarget 2018; 8:2536-2549. [PMID: 27911860 PMCID: PMC5356822 DOI: 10.18632/oncotarget.13701] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/21/2016] [Indexed: 12/12/2022] Open
Abstract
Given that mutated p53 (50% of all human cancers) is over-expressed in many cancers, restoration of mutant p53 to its wild type biological function has been sought after as cancer therapy. The conformational flexibility has allowed to restore the normal biological function of mutant p53 by short peptides and small molecule compounds. Recently, studies have focused on physiological mechanisms such as acetylation of lysine residues to rescue the wild type activity of mutant p53. Using p53 null prostate cancer cell line we show that ID4 dependent acetylation promotes mutant p53 DNA-binding capabilities to its wild type consensus sequence, thus regulating p53-dependent target genes leading to subsequent cell cycle arrest and apoptosis. Specifically, by using wild type, mutant (P223L, V274F, R175H, R273H), acetylation mimics (K320Q and K373Q) and non-acetylation mimics (K320R and K373R) of p53, we identify that ID4 promotes acetylation of K373 and to a lesser extent K320, in turn restoring p53-dependent biological activities. Together, our data provides a molecular understanding of ID4 dependent acetylation that suggests a strategy of enhancing p53 acetylation at sites K373 and K320 that may serve as a viable mechanism of physiological restoration of mutant p53 to its wild type biological function.
Collapse
|
24
|
Lee J, Kim Y, Liu T, Hwang YJ, Hyeon SJ, Im H, Lee K, Alvarez VE, McKee AC, Um SJ, Hur M, Mook-Jung I, Kowall NW, Ryu H. SIRT3 deregulation is linked to mitochondrial dysfunction in Alzheimer's disease. Aging Cell 2018; 17. [PMID: 29130578 PMCID: PMC5771400 DOI: 10.1111/acel.12679] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2017] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly. Despite decades of study, effective treatments for AD are lacking. Mitochondrial dysfunction has been closely linked to the pathogenesis of AD, but the relationship between mitochondrial pathology and neuronal damage is poorly understood. Sirtuins (SIRT, silent mating type information regulation 2 homolog in yeast) are NAD-dependent histone deacetylases involved in aging and longevity. The objective of this study was to investigate the relationship between SIRT3 and mitochondrial function and neuronal activity in AD. SIRT3 mRNA and protein levels were significantly decreased in AD cerebral cortex, and Ac-p53 K320 was significantly increased in AD mitochondria. SIRT3 prevented p53-induced mitochondrial dysfunction and neuronal damage in a deacetylase activity-dependent manner. Notably, mitochondrially targeted p53 (mito-p53) directly reduced mitochondria DNA-encoded ND2 and ND4 gene expression resulting in increased reactive oxygen species (ROS) and reduced mitochondrial oxygen consumption. ND2 and ND4 gene expressions were significantly decreased in patients with AD. p53-ChIP analysis verified the presence of p53-binding elements in the human mitochondrial genome and increased p53 occupancy of mitochondrial DNA in AD. SIRT3 overexpression restored the expression of ND2 and ND4 and improved mitochondrial oxygen consumption by repressing mito-p53 activity. Our results indicate that SIRT3 dysfunction leads to p53-mediated mitochondrial and neuronal damage in AD. Therapeutic modulation of SIRT3 activity may ameliorate mitochondrial pathology and neurodegeneration in AD.
Collapse
Affiliation(s)
- Junghee Lee
- VA Boston Healthcare System; Boston MA 02130 USA
- Alzheimer's Disease Center and Department of Neurology; Boston University School of Medicine; Boston MA 02118 USA
| | - Yunha Kim
- Laboratory for Neuronal Gene Regulation and Epigenetics; Center for NeuroMedicine; Brain Science Institute; Korea Institute of Science and Technology; Seoul 02792 South Korea
| | - Tian Liu
- Laboratory for Neuronal Gene Regulation and Epigenetics; Center for NeuroMedicine; Brain Science Institute; Korea Institute of Science and Technology; Seoul 02792 South Korea
| | - Yu Jin Hwang
- Laboratory for Neuronal Gene Regulation and Epigenetics; Center for NeuroMedicine; Brain Science Institute; Korea Institute of Science and Technology; Seoul 02792 South Korea
| | - Seung Jae Hyeon
- Laboratory for Neuronal Gene Regulation and Epigenetics; Center for NeuroMedicine; Brain Science Institute; Korea Institute of Science and Technology; Seoul 02792 South Korea
| | - Hyeonjoo Im
- Laboratory for Neuronal Gene Regulation and Epigenetics; Center for NeuroMedicine; Brain Science Institute; Korea Institute of Science and Technology; Seoul 02792 South Korea
| | - Kyungeun Lee
- Advanced Analysis Center; Korea Institute of Science and Technology; Seoul 02792 South Korea
| | - Victor E. Alvarez
- Alzheimer's Disease Center and Department of Neurology; Boston University School of Medicine; Boston MA 02118 USA
- Bedford VA Medical Center; Bedford MA 01730 USA
| | - Ann C. McKee
- VA Boston Healthcare System; Boston MA 02130 USA
- Alzheimer's Disease Center and Department of Neurology; Boston University School of Medicine; Boston MA 02118 USA
| | - Soo-Jong Um
- Department of Integrative Bioscience and Biotechnology; Sejong University; Seoul 05006 South Korea
| | - Manwook Hur
- Department of Biochemistry; Yonsei University College of Medicine; Seoul 03722 South Korea
| | - Inhee Mook-Jung
- Departments of Biochemistry and Biomedical Sciences; Seoul National University College of Medicine; Seoul 03080 South Korea
| | - Neil W. Kowall
- VA Boston Healthcare System; Boston MA 02130 USA
- Alzheimer's Disease Center and Department of Neurology; Boston University School of Medicine; Boston MA 02118 USA
| | - Hoon Ryu
- VA Boston Healthcare System; Boston MA 02130 USA
- Alzheimer's Disease Center and Department of Neurology; Boston University School of Medicine; Boston MA 02118 USA
- Laboratory for Neuronal Gene Regulation and Epigenetics; Center for NeuroMedicine; Brain Science Institute; Korea Institute of Science and Technology; Seoul 02792 South Korea
| |
Collapse
|
25
|
Balliu M, Cellai C, Lulli M, Laurenzana A, Torre E, Vannucchi AM, Paoletti F. HDAC1 controls CIP2A transcription in human colorectal cancer cells. Oncotarget 2017; 7:25862-71. [PMID: 27029072 PMCID: PMC5041950 DOI: 10.18632/oncotarget.8406] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/10/2016] [Indexed: 12/03/2022] Open
Abstract
This work describes the effectiveness of HDAC-inhibitor (S)-2 towards colorectal cancer (CRC) HCT116 cells in vitro by inducing cell cycle arrest and apoptosis, and in vivo by contrasting tumour growth in mice xenografts. Among the multifaceted drug-induced events described herein, an interesting link has emerged between the oncoprotein histone deacetylase HDAC1 and the oncogenic Cancerous Inhibitor of Protein Phosphatase 2A (CIP2A) which is overexpressed in several cancers including CRCs. HDAC1 inhibition by (S)-2 or specific siRNAs downregulates CIP2A transcription in three different CRC cell lines, thus restoring the oncosuppressor phosphatase PP2A activity that is reduced in most cancers. Once re-activated, PP2A dephosphorylates pGSK-3β(ser9) which phosphorylates β-catenin that remains within the cytosol where it undergoes degradation. The decreased amount/activity of β-catenin transcription factor prompts cell growth arrest by diminishing c-Myc and cyclin D1 expression and abrogating the prosurvival Wnt/β-catenin signaling pathway. These results are the first evidence that the inhibition of HDAC1 by (S)-2 downregulates CIP2A transcription and unleashes PP2A activity, thus inducing growth arrest and apoptosis in CRC cells.
Collapse
Affiliation(s)
- Manjola Balliu
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy
| | - Cristina Cellai
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", 50134 Firenze, Italy
| | - Matteo Lulli
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", 50134 Firenze, Italy
| | - Anna Laurenzana
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", 50134 Firenze, Italy
| | - Eugenio Torre
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", 50134 Firenze, Italy
| | | | - Francesco Paoletti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", 50134 Firenze, Italy
| |
Collapse
|
26
|
Mekala JR, Naushad SM, Ponnusamy L, Arivazhagan G, Sakthiprasad V, Pal-Bhadra M. Epigenetic regulation of miR-200 as the potential strategy for the therapy against triple-negative breast cancer. Gene 2017; 641:248-258. [PMID: 29038000 DOI: 10.1016/j.gene.2017.10.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/15/2017] [Accepted: 10/07/2017] [Indexed: 02/08/2023]
Abstract
MicroRNAs (miRNAs) are a class of small, non-coding RNAs that are involved in the regulation of gene expression at the post-transcriptional level. MicroRNAs play an important role in cancer cell proliferation, survival and apoptosis. Epigenetic modifiers regulate the microRNA expression. Among the epigenetic players, histone deacetylases (HDACs) function as the key regulators of microRNA expression. Epigenetic machineries such as DNA and histone modifying enzymes and various microRNAs have been identified as the important contributors in cancer initiation and progression. Recent studies have shown that developing innovative microRNA-targeting therapies might improve the human health, specifically against the disease areas of high unmet medical need. Thus microRNA based therapeutics are gaining importance for anti-cancer therapy. Studies on Triple negative breast cancer (TNBC) have revealed the early relapse and poor overall survival of patients which needs immediate therapeutic attention. In this report, we focus the effect of HDAC inhibitors on TNBC cell proliferation, regulation of microRNA gene expression by a series of HDAC genes, chromatin epigenetics, epigenetic remodelling at miR-200 promoter and its modulation by various HDACs. We also discuss the need for identifying novel HDAC inhibitors for modulation of miR-200 in triple negative breast cancer.
Collapse
Affiliation(s)
- Janaki Ramaiah Mekala
- School of Chemical and Biotechnology, SASTRA University, Tirumalaisamudram, Thanjavur 613401, India.
| | | | - Lavanya Ponnusamy
- School of Chemical and Biotechnology, SASTRA University, Tirumalaisamudram, Thanjavur 613401, India
| | - Gayatri Arivazhagan
- School of Chemical and Biotechnology, SASTRA University, Tirumalaisamudram, Thanjavur 613401, India
| | - Vaishnave Sakthiprasad
- School of Chemical and Biotechnology, SASTRA University, Tirumalaisamudram, Thanjavur 613401, India
| | - Manika Pal-Bhadra
- CSIR - Centre for Chemical Biology, CSIR-IICT, Hyderabad 500007, Telangana, India
| |
Collapse
|
27
|
Milstone ZJ, Lawson G, Trivedi CM. Histone deacetylase 1 and 2 are essential for murine neural crest proliferation, pharyngeal arch development, and craniofacial morphogenesis. Dev Dyn 2017; 246:1015-1026. [PMID: 28791750 DOI: 10.1002/dvdy.24563] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 08/02/2017] [Accepted: 08/07/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Craniofacial anomalies involve defective pharyngeal arch development and neural crest function. Copy number variation at 1p35, containing histone deacetylase 1 (Hdac1), or 6q21-22, containing Hdac2, are implicated in patients with craniofacial defects, suggesting an important role in guiding neural crest development. However, the roles of Hdac1 and Hdac2 within neural crest cells remain unknown. RESULTS The neural crest and its derivatives express both Hdac1 and Hdac2 during early murine development. Ablation of Hdac1 and Hdac2 within murine neural crest progenitor cells cause severe hemorrhage, atrophic pharyngeal arches, defective head morphogenesis, and complete embryonic lethality. Embryos lacking Hdac1 and Hdac2 in the neural crest exhibit decreased proliferation and increased apoptosis in both the neural tube and the first pharyngeal arch. Mechanistically, loss of Hdac1 and Hdac2 upregulates cyclin-dependent kinase inhibitors Cdkn1a, Cdkn1b, Cdkn1c, Cdkn2b, Cdkn2c, and Tp53 within the first pharyngeal arch. CONCLUSIONS Our results show that Hdac1 and Hdac2 function redundantly within the neural crest to regulate proliferation and the development of the pharyngeal arches by means of repression of cyclin-dependent kinase inhibitors. Developmental Dynamics 246:1015-1026, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Zachary J Milstone
- Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.,Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Grace Lawson
- Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Chinmay M Trivedi
- Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.,Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.,Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
28
|
Cao GD, Chen B, Xiong MM. Role of metastasis-associated protein 1 in prognosis of patients with digestive tract cancers: A meta-analysis. PLoS One 2017; 12:e0176431. [PMID: 28570554 PMCID: PMC5453427 DOI: 10.1371/journal.pone.0176431] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 04/09/2017] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES Metastasis-associated protein 1 (MTA1) is a transcriptional regulator and significantly associated with prognosis of patients with cancer. However, its role as a potential prognostic marker in digestive tract cancer (DTC) is controversial. In this study, a meta-analysis was conducted to evaluate the MTA1 expression as a predictor of clinicopathology and survival of patients with DTC. METHODS We searched PubMed, Ovid, Web of Science and Cochrane databases using multiple search strategies for eligible studies. STATA 11.0 software was used to pool the data and analyze the association, odds ratios (ORs) and 95% confidence intervals (CIs) were used to measure the strength of the association. Furthermore, the Newcastle-Ottawa scale was used to evaluate the quality of eligible studies. RESULTS MTA1 overexpression was strongly associated with depth of invasion (OR = 1.88, 95%CI: 1.05-3.37, P = 0.03), lymph node metastasis (OR = 2.30, 95%CI: 1.76-3.01, P<0.001), vascular invasion (OR = 2.02, 95%CI: 1.40-2.91, P<0.001) and TNM stage (OR = 2.78, 95%CI: 1.63-4.74, P<0.001), and was related to 1- (RR = 1.84, 95%CI: 1.18-2.89, P = 0.008), 3- (RR = 1.74, 95%CI: 1.32-2.30, P<0.001) and 5-year (RR = 1.64, 95%CI: 1.18-2.27, P = 0.003) OS. Further, MTA1 was associated with 1- (RR = 4.16, 95%CI: 1.35-12.81, P = 0.01), 3- (RR = 1.90, 95%CI: 1.02-3.53, P = 0.04) and 5- (RR = 2.17, 95%CI: 1.41-3.32, P<0.001) year DFS. In subgroup analyses based on study quality and tumor type, MTA1 overexpression was obviously related to clinical parameters, such as lymph node metastasis and TNM stage, and was also associated with prognosis of patients with gastrointestinal or esophageal cancer. CONCLUSIONS MTA1 expression is strongly correlated with metastasis-related variables, and represents a promising prognostic factor in DTC.
Collapse
Affiliation(s)
| | - Bo Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mao-ming Xiong
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
29
|
Parbin S, Shilpi A, Kar S, Pradhan N, Sengupta D, Deb M, Rath SK, Patra SK. Insights into the molecular interactions of thymoquinone with histone deacetylase: evaluation of the therapeutic intervention potential against breast cancer. MOLECULAR BIOSYSTEMS 2016; 12:48-58. [PMID: 26540192 DOI: 10.1039/c5mb00412h] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Many HDAC inhibitors have passed through the gateway of clinical trials. However, they have limited therapeutic implications due to their pleiotropic pharmaceutical properties and off-target effects. In view of this, dietary active phytochemicals were evaluated. Based upon the chemical and structural insights of HDAC active pockets, thymoquinone (TQ) was investigated to uncover its active participation in HDAC inhibition. The synergistic analysis of docking and molecular dynamics simulation disclosed the elementary interaction and stability of TQ with human HDACs. The in silico findings were corroborated with an in vitro analysis, demonstrating the efficient role of TQ in the attenuation of global HDAC activity. Furthermore, TQ also elicited downstream effects of HDAC inhibition: reactivation of HDAC target genes (p21 and Maspin), induction of the pro-apoptotic gene Bax, down regulation of the anti-apoptotic gene Bcl-2 and arrest of the cell cycle at the G2/M phase. Finally, the result of a higher cytotoxicity of TQ towards MCF-7 breast cancer cells in comparison to normal cells indicates the potential of TQ to be an anticancer drug.
Collapse
Affiliation(s)
- Sabnam Parbin
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha-769008, India.
| | - Arunima Shilpi
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha-769008, India.
| | - Swayamsiddha Kar
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha-769008, India.
| | - Nibedita Pradhan
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha-769008, India.
| | - Dipta Sengupta
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha-769008, India.
| | - Moonmoon Deb
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha-769008, India.
| | - Sandip Kumar Rath
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha-769008, India.
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha-769008, India.
| |
Collapse
|
30
|
YANG HAIPING, XU LIJUAN, QIAN HAILI, NIU XINQIANG, ZHAO DAN, ZHAO ZHILONG, WU JUN, LIU JUNFENG, WANG YANYU. Correlation between insulin‑like growth factor binding protein 3 and metastasis‑associated gene 1 protein in esophageal squamous cell carcinoma. Mol Med Rep 2016; 13:4143-50. [PMID: 27035126 PMCID: PMC4838119 DOI: 10.3892/mmr.2016.5046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 11/25/2015] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to investigate the correlation between insulin‑like growth factor binding protein 3 (IGFBP‑3) and metastasis‑associated gene 1 (MTA1) protein, and the clinicopathological features and prognosis of esophageal squamous cell carcinoma (ESCC). Patients with ESCC who underwent surgical resection were enrolled in the current study, ESCC tissues and adjacent normal tissues (control) were obtained from 197 patients. The protein expression levels of IGFBP‑3 and MTA1 were detected using immunohistochemistry. The results demonstrated that the expression of IGFBP‑3 in ESCC tissues was significantly lower than in the adjacent normal tissues (27.4 vs. 40.6%; P<0.05), and was negatively correlated with smoking status, degree of tumor differentiation and lymph node metastasis (P<0.05). The expression of MTA1 protein in ESCC tissues was significantly higher than that of the adjacent tissues (42.1 vs. 11.2%; P<0.05), and was positively correlated with the tumor size, extent of tumor invasion and lymph node metastasis (P<0.05). No association was identified between the protein expression levels of IGFBP‑3 and MTA1. The protein expression levels of IGFBP‑3 and MTA1 were not independent risk factors for ESCC prognosis; however, the degree of tumor invasion (P=0.02) and rate of lymph node metastasis (P=0.027) were. IGFBP‑3 inhibits the proliferation and metastasis of ESCC; however, MTA1 promotes the proliferation and metastasis of ESCC. There is no interaction between IGFBP‑3 and MTA1 in ESCC, and they are not independent risk factors for ESCC prognosis.
Collapse
Affiliation(s)
- HAIPING YANG
- Department of Thoracic and Cardiovascular Surgery, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing 101100, P.R. China
| | - LIJUAN XU
- Department of Thoracic and Cardiovascular Surgery, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing 101100, P.R. China
| | - HAILI QIAN
- State Key Laboratory of Molecular Oncology, Cancer Hospital/Institute, Chinese Academy of Medical Sciences, Beijing 100021, P.R. China
| | - XINQIANG NIU
- Department of Thoracic Surgery, Cixian People's Hospital, Handan, Hebei 056500, P.R. China
| | - DAN ZHAO
- Department of Pathology, Beijing Chest Hospital of Capital Medical University, Beijing 101100, P.R. China
| | - ZHILONG ZHAO
- Department of Cardiothoracics, Zhongshan Hospital, Dalian University, Dalian, Liaoning 116001, P.R. China
| | - JUN WU
- Department of Thoracic and Cardiovascular Surgery, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing 101100, P.R. China
| | - JUNFENG LIU
- Department of Thoracic Surgery, Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - YANYU WANG
- Department of Thoracic and Cardiovascular Surgery, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing 101100, P.R. China
| |
Collapse
|
31
|
Liu YC, Chang PY, Chao CCK. CITED2 silencing sensitizes cancer cells to cisplatin by inhibiting p53 trans-activation and chromatin relaxation on the ERCC1 DNA repair gene. Nucleic Acids Res 2015; 43:10760-81. [PMID: 26384430 PMCID: PMC4678856 DOI: 10.1093/nar/gkv934] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/08/2015] [Indexed: 02/07/2023] Open
Abstract
In this study, we show that silencing of CITED2 using small-hairpin RNA (shCITED2) induced DNA damage and reduction of ERCC1 gene expression in HEK293, HeLa and H1299 cells, even in the absence of cisplatin. In contrast, ectopic expression of ERCC1 significantly reduced intrinsic and induced DNA damage levels, and rescued the effects of CITED2 silencing on cell viability. The effects of CITED2 silencing on DNA repair and cell death were associated with p53 activity. Furthermore, CITED2 silencing caused severe elimination of the p300 protein and markers of relaxed chromatin (acetylated H3 and H4, i.e. H3K9Ac and H3K14Ac) in HEK293 cells. Chromatin immunoprecipitation assays further revealed that DNA damage induced binding of p53 along with H3K9Ac or H3K14Ac at the ERCC1 promoter, an effect which was almost entirely abrogated by silencing of CITED2 or p300. Moreover, lentivirus-based CITED2 silencing sensitized HeLa cell line-derived tumor xenografts to cisplatin in immune-deficient mice. These results demonstrate that CITED2/p300 can be recruited by p53 at the promoter of the repair gene ERCC1 in response to cisplatin-induced DNA damage. The CITED2/p300/p53/ERCC1 pathway is thus involved in the cell response to cisplatin and represents a potential target for cancer therapy.
Collapse
Affiliation(s)
- Yu-Chin Liu
- Tumor Biology Laboratory, Department of Biochemistry and Molecular Biology, Chang Gung University, 259 Wen-Hua first Road, Gueishan, Taoyuan 333, Taiwan, Republic of China Graduate Institute of Biomedical Sciences, Chang Gung University, 259 Wen-Hua first Road, Gueishan,Taoyuan 333, Taiwan, Republic of China
| | - Pu-Yuan Chang
- Tumor Biology Laboratory, Department of Biochemistry and Molecular Biology, Chang Gung University, 259 Wen-Hua first Road, Gueishan, Taoyuan 333, Taiwan, Republic of China
| | - Chuck C-K Chao
- Tumor Biology Laboratory, Department of Biochemistry and Molecular Biology, Chang Gung University, 259 Wen-Hua first Road, Gueishan, Taoyuan 333, Taiwan, Republic of China Graduate Institute of Biomedical Sciences, Chang Gung University, 259 Wen-Hua first Road, Gueishan,Taoyuan 333, Taiwan, Republic of China
| |
Collapse
|
32
|
Ali SR, Humphreys KJ, McKinnon RA, Michael MZ. Impact of Histone Deacetylase Inhibitors on microRNA Expression and Cancer Therapy: A Review. Drug Dev Res 2015; 76:296-317. [PMID: 26303212 DOI: 10.1002/ddr.21268] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chromatin-modifying drugs, such as histone deacetylase inhibitors (HDACi), have shown potential as cancer therapeutics, either alone or in combination with other therapies. HDACi have the ability to reverse aberrant epigenetic modifications associated with cancer, namely dysregulated histone acetylation. There are currently three FDA approved HDACi; vorinostat, romidepsin, and panobinostat. Epigenetic modifications can regulate the expression of protein coding genes, and in addition can alter expression of microRNA (miRNA) genes. Many miRNAs play key roles in cell proliferation and apoptosis, and are commonly dysregulated in cancer states. A number of in vitro and in vivo studies have demonstrated the ability of chromatin-modifying drugs to alter miRNA expression, which may provide the basis for further investigation of miRNAs as therapeutic targets or as biomarkers of drug response. This review summarises findings from studies investigating the effects of HDACi on miRNA expression, as well as key clinical trials involving HDACi. Understanding how chromatin-modifying drugs epigenetically modulate miRNA genes provides further insight into the cellular mechanisms that deliver therapeutic responses, and may assist in refining treatment strategies.
Collapse
Affiliation(s)
- Saira R Ali
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Karen J Humphreys
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Ross A McKinnon
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Michael Z Michael
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, South Australia, Australia.,Department of Gastroenterology and Hepatology, Flinders Medical Centre, Adelaide, South Australia, Australia
| |
Collapse
|
33
|
Stengel KR, Hiebert SW. Class I HDACs Affect DNA Replication, Repair, and Chromatin Structure: Implications for Cancer Therapy. Antioxid Redox Signal 2015; 23:51-65. [PMID: 24730655 PMCID: PMC4492608 DOI: 10.1089/ars.2014.5915] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE The contribution of epigenetic alterations to cancer development and progression is becoming increasingly clear, prompting the development of epigenetic therapies. Histone deacetylase inhibitors (HDIs) represent one of the first classes of such therapy. Two HDIs, Vorinostat and Romidepsin, are broad-spectrum inhibitors that target multiple histone deacetylases (HDACs) and are FDA approved for the treatment of cutaneous T-cell lymphoma. However, the mechanism of action and the basis for the cancer-selective effects of these inhibitors are still unclear. RECENT ADVANCES While the anti-tumor effects of HDIs have traditionally been attributed to their ability to modify gene expression after the accumulation of histone acetylation, recent studies have identified the effects of HDACs on DNA replication, DNA repair, and genome stability. In addition, the HDIs available in the clinic target multiple HDACs, making it difficult to assign either their anti-tumor effects or their associated toxicities to the inhibition of a single protein. However, recent studies in mouse models provide insights into the tissue-specific functions of individual HDACs and their involvement in mediating the effects of HDI therapy. CRITICAL ISSUES Here, we describe how altered replication contributes to the efficacy of HDAC-targeted therapies as well as discuss what knowledge mouse models have provided to our understanding of the specific functions of class I HDACs, their potential involvement in tumorigenesis, and how their disruption may contribute to toxicities associated with HDI treatment. FUTURE DIRECTIONS Impairment of DNA replication by HDIs has important therapeutic implications. Future studies should assess how best to exploit these findings for therapeutic gain.
Collapse
Affiliation(s)
- Kristy R. Stengel
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Scott W. Hiebert
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
34
|
Ngollo M, Dagdemir A, Karsli-Ceppioglu S, Judes G, Pajon A, Penault-Llorca F, Boiteux JP, Bignon YJ, Guy L, Bernard-Gallon DJ. Epigenetic modifications in prostate cancer. Epigenomics 2015; 6:415-26. [PMID: 25333850 DOI: 10.2217/epi.14.34] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer is the most common cancer in men and the second leading cause of cancer deaths in men in France. Apart from the genetic alterations in prostate cancer, epigenetics modifications are involved in the development and progression of this disease. Epigenetic events are the main cause in gene regulation and the three most epigenetic mechanisms studied include DNA methylation, histone modifications and microRNA expression. In this review, we summarized epigenetic mechanisms in prostate cancer. Epigenetic drugs that inhibit DNA methylation, histone methylation and histone acetylation might be able to reactivate silenced gene expression in prostate cancer. However, further understanding of interactions of these enzymes and their effects on transcription regulation in prostate cancer is needed and has become a priority in biomedical research. In this study, we summed up epigenetic changes with emphasis on pharmacologic epigenetic target agents.
Collapse
Affiliation(s)
- Marjolaine Ngollo
- Department of Oncogenetics, Centre Jean Perrin, CBRV, 28 place Henri Dunant, BP 38, 63001 Clermont-Ferrand, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Urbinati G, Marsaud V, Nicolas V, Vergnaud-Gauduchon J, Renoir JM. Liposomal trichostatin A: therapeutic potential in hormone-dependent and -independent breast cancer xenograft models. Horm Mol Biol Clin Investig 2015; 6:215-25. [PMID: 25961258 DOI: 10.1515/hmbci.2011.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 01/12/2011] [Indexed: 12/31/2022]
Abstract
BACKGROUND Trichostatin A (TSA) is one of the most potent histone deacetylase inhibitors (HDACi) in vitro but it lacks biological activity in vivo when injected intravenously owing to its fast metabolism. MATERIALS AND METHODS TSA was incorporated into Stealth® liposomes (TSA-lipo) at a high loading and its anticancer activity was evaluated in several types of breast cancer cells and xenografts. RESULTS In estrogen receptor α (ERα)-positive MCF-7 and T47-D cells, TSA induced a long-term degradation of cyclin A and a proteasome-dependent loss of ERα and cyclin D1, allowed derepression of p21WAF1/CIP1, HDAC1 and RhoB GTPase, concomitantly with blockade in G2/M of the cell cycle and apoptosis induction. In MDA-MB-231 (MDA) and SKBr-3 cells, TSA increased ERα mRNA and p21WAF1/CIP1 protein expression, but decreased cyclin A with a G2/M blockade and cleavage of polyADP-ribose polymerase (PARP). No significant restoration of any ER protein was noticed in any cells. TSA-lipo markedly inhibited tumor growth in MCF-7 and MDA cells xenografts following intravenous injection. Their anticancer effects were characterized by inhibition of Ki-67 labeling, the inhibition of tumor vasculature and an increase of p21WAF1/CIP1 in both tumors. In MCF-7 cell tumors, enhanced RhoB accumulation in the cytoplasm of epithelial cells was noticed, inversely to ERα that was strongly decreased. CONCLUSION Such anticancer activity of TSA-lipo is exp-lained by the protection provided by HDACi encapsulation and by the strong tumor accumulation of the nanocarriers as revealed by fluorescence confocal microscopy experi-ments. Together with its lack of toxicity, the enhanced stability of TSA-lipo in vivo justifies its development for therapeutic use in the treatment estradiol-dependent and -independent breast cancers.
Collapse
|
36
|
Chatterjee N, Tenniswood M. The potential of histone deacetylase inhibitors in breast cancer therapy. BREAST CANCER MANAGEMENT 2015. [DOI: 10.2217/bmt.14.56] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Breast cancer is the second leading cause of cancer-related mortality in women. Despite improvements in prevention, detection and treatment, breast cancer will be responsible for nearly 40,000 deaths in 2014. The function of histone deacetylases (HDACs) and their potential as therapeutic targets has become an area of intense investigation and small molecule inhibitors of HDACs (HDACi) are now being investigated as potential chemotherapeutics for breast cancer. In addition to altering chromatin structure through stabilization of histone acetylation, HDACi treatment induces the accumulation of acetylated isoforms of many nonhistone proteins, altering their structure and function. These structural changes influence protein–protein interactions and cellular processes including cell cycle arrest, apoptosis, autophagy, induction of reactive oxygen species and mitotic catastrophe. While the usefulness of these compounds as single agents for treatment of breast cancer is still under investigation, cotreatment with other therapies is being evaluated in a number of clinical trials.
Collapse
Affiliation(s)
- Namita Chatterjee
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, 1 Discovery Drive, Rensselaer, NY 12144, USA
| | - Martin Tenniswood
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, 1 Discovery Drive, Rensselaer, NY 12144, USA
| |
Collapse
|
37
|
Kim JS, Lee SC, Min HY, Park KH, Hyun SY, Kwon SJ, Choi SP, Kim WY, Lee HJ, Lee HY. Activation of insulin-like growth factor receptor signaling mediates resistance to histone deacetylase inhibitors. Cancer Lett 2015; 361:197-206. [PMID: 25721083 DOI: 10.1016/j.canlet.2015.02.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 02/16/2015] [Accepted: 02/16/2015] [Indexed: 11/30/2022]
Abstract
Histone deacetylases (HDACs) are considered promising targets in the treatment of hematologic malignancies and several types of solid tumors, including non-small cell lung cancer (NSCLC). However, the efficacy of HDAC inhibitors in solid tumors is marginal, and the mechanisms underlying resistance to HDAC inhibitors are largely unknown. Here, we demonstrate the involvement of type 1 insulin-like growth factor receptor (IGF-1R) signaling in resistance to HDAC inhibitors in NSCLC. Using MTT and soft-agar colony formation assays, we selected NSCLC cell lines that exhibited intrinsic resistance to vorinostat. Treatment with vorinostat activated IGF-1R signaling in vorinostat-resistant but not vorinostat-sensitive NSCLC cells. Other HDAC inhibitors, including trichostatin A, sodium butyrate, and depsipeptide, also activated IGF-1R signaling in vorinostat-resistant NSCLC cells. Blockade of IGF-1R signaling via IGF-1R monoclonal antibodies (mAbs) or through knockdown of IGF-1R via RNA interference sensitized vorinostat-resistant cells to HDAC inhibition. Finally, IGF-1R mAbs sensitized xenograft tumors of vorinostat-resistant cells to vorinostat treatment in vivo. These findings suggest that IGF-1R activation is generally involved in resistance to HDAC inhibitors and that targeting IGF-1R is an effective strategy for overcoming resistance to HDAC inhibitors in NSCLC.
Collapse
Affiliation(s)
- Jin-Soo Kim
- Department of Thoracic Head & Neck Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Su-Chan Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Hye-Young Min
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Kwan Hee Park
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Seung Yeob Hyun
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - So Jung Kwon
- College of Pharmacy, Inje University, Gimhae, Gyungnam 621-749, Republic of Korea
| | - Sun Phil Choi
- Department of Thoracic Head & Neck Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Woo-Young Kim
- Department of Thoracic Head & Neck Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Hyo-Jong Lee
- College of Pharmacy, Inje University, Gimhae, Gyungnam 621-749, Republic of Korea
| | - Ho-Young Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|
38
|
Singh R, Pandey PN. Molecular docking and molecular dynamics study on SmHDAC1 to identify potential lead compounds against Schistosomiasis. Mol Biol Rep 2015; 42:689-98. [DOI: 10.1007/s11033-014-3816-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 11/04/2014] [Indexed: 10/24/2022]
|
39
|
Chu QSC, Nielsen TO, Alcindor T, Gupta A, Endo M, Goytain A, Xu H, Verma S, Tozer R, Knowling M, Bramwell VB, Powers J, Seymour LK, Eisenhauer EA. A phase II study of SB939, a novel pan-histone deacetylase inhibitor, in patients with translocation-associated recurrent/metastatic sarcomas-NCIC-CTG IND 200†. Ann Oncol 2015; 26:973-981. [PMID: 25632070 DOI: 10.1093/annonc/mdv033] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 01/09/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND A subgroup of sarcomas is characterized by defining chromosomal translocations, creating fusion transcription factor oncogenes. Resultant fusion oncoproteins associate with chromatin-modifying complexes containing histone deacetylases (HDAC), and lead to epigenetic transcriptional dysregulation. HDAC inhibitors were shown to be effective in vitro, reversing gene repression by these complexes, restoring PTEN expression and apoptosis via the PI3K/Akt/mTOR pathway. PATIENTS AND METHODS SB939 is an oral inhibitor of classes 1 and 2 HDAC. Eligible patients with recurrent or metastatic translocation-associated sarcoma (TAS) by local pathology were treated with 60 mg/day every other day for 3 of 4 weeks. Central pathology review was conducted with fusion oncogenes characterized, and HDAC2 expression correlated with efficacy in pre-specified methods. RESULTS Twenty-two patients were treated with a median of 2 cycles. Fourteen patients were assessable for response with confirmed specific chromosomal translocations; 8 had a best response of stable disease (SD) (median duration 5.4 months) with no confirmed objective responses. The 3-month progression-free survival (PFS) rate was 49%. Among those with HDAC2 score ≥5, 7/10 had SD, versus 0/3 with HDAC2 score <5. SB939 was considered as well tolerated with <10% patients experienced ≥grade 3 toxicity. CONCLUSION This study was stopped prematurely due to prolonged unavailability of SB939. No objective responses were seen. Although the observed SD in HDAC2 high patients was interesting, due to the small sample size, no definitive conclusion can be drawn about the efficacy of SB939 in this patient population. CLINICAL TRIAL NCT01112384.
Collapse
Affiliation(s)
- Q S-C Chu
- Division of Medical Oncology, Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton.
| | - T O Nielsen
- Department of Pathology, British Columbia Cancer Agency, Vancouver
| | - T Alcindor
- Division of Medical Oncology, Department of Oncology, McGill University, Montreal
| | - A Gupta
- Division of Medical Oncology, Department of Oncology, Mount Sinai Hospital/Princess Margaret Hospital, University of Toronto, Toronto, Canada
| | - M Endo
- Department of Orthopedic Surgery, Kyushu University, Fukuoka, Japan
| | - A Goytain
- Department of Pathology, British Columbia Cancer Agency, Vancouver
| | - H Xu
- Investigational New Drug Program, NCIC-Clinical Trials Group, Kingston
| | - S Verma
- Department of Medical Oncology, Ottawa Cancer Centre, University of Ottawa, Ottawa
| | - R Tozer
- Division of Medical Oncology, Department of Oncology, Jurvinski Cancer Centre, McMaster University, Hamilton
| | - M Knowling
- Department of Pathology, British Columbia Cancer Agency, Vancouver
| | - V B Bramwell
- Division of Medical Oncology, Department of Oncology, Tom Baker Cancer Centre, University of Calgary, Calgary, Canada
| | - J Powers
- Investigational New Drug Program, NCIC-Clinical Trials Group, Kingston
| | - L K Seymour
- Investigational New Drug Program, NCIC-Clinical Trials Group, Kingston
| | - E A Eisenhauer
- Investigational New Drug Program, NCIC-Clinical Trials Group, Kingston
| |
Collapse
|
40
|
Huang Z, Peng S, Knoff J, Lee SY, Yang B, Wu TC, Hung CF. Combination of proteasome and HDAC inhibitor enhances HPV16 E7-specific CD8+ T cell immune response and antitumor effects in a preclinical cervical cancer model. J Biomed Sci 2015; 22:7. [PMID: 25591912 PMCID: PMC4298946 DOI: 10.1186/s12929-014-0111-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 12/31/2014] [Indexed: 11/15/2022] Open
Abstract
Background Bortezomib, a proteasome inhibitor and suberoylanilide hydroxamic acid (SAHA, also known as Vorinostat), a histone deacetylase inhibitor, have been recognized as potent chemotherapeutic drugs. Bortezomib and SAHA are FDA-approved for the treatment of cutaneous T cell lymphoma and multiple myeloma/mantle cell lymphoma, respectively. Furthermore, the combination of the bortezomib and SAHA has been tested in a variety of preclinical models and in clinical trials and may be ideal for the treatment of cancer. However, it remains unclear how this treatment strategy affects the host immune response against tumors. Results Here, we used a well-defined E6/E7-expressing tumor model to examine how the immune system can be motivated to act against tumor cells expressing tumor antigens. We demonstrate that the combination of bortezomib and SAHA elicits potent antitumor effects in TC-1 tumor-bearing mice. Additionally, we are the first to show that treatment with bortezomib and SAHA leads to tumor-specific immunity by rendering tumor cells more susceptible to killing by antigen-specific CD8+ T cells than treatment with either drug alone. Conclusions The current study serves an important foundation for the future clinical application of both drugs for the treatment of cervical cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12929-014-0111-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhuomin Huang
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA. .,Department of Gynecology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China.
| | - Shiwen Peng
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA.
| | - Jayne Knoff
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA.
| | - Sung Yong Lee
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA. .,Department of Internal Medicine, Korea University Medical Center, Seoul, South Korea.
| | - Benjamin Yang
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA.
| | - Tzyy-Choou Wu
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA. .,Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, MD, USA. .,Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions, Baltimore, MD, USA. .,Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA.
| |
Collapse
|
41
|
Zhang J, Zhong Q. Histone deacetylase inhibitors and cell death. Cell Mol Life Sci 2014; 71:3885-901. [PMID: 24898083 PMCID: PMC4414051 DOI: 10.1007/s00018-014-1656-6] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 04/23/2014] [Accepted: 05/20/2014] [Indexed: 12/14/2022]
Abstract
Histone deacetylases (HDACs) are a vast family of enzymes involved in chromatin remodeling and have crucial roles in numerous biological processes, largely through their repressive influence on transcription. In addition to modifying histones, HDACs also target many other non-histone protein substrates to regulate gene expression. Recently, HDACs have gained growing attention as HDAC-inhibiting compounds are being developed as promising cancer therapeutics. Histone deacetylase inhibitors (HDACi) have been shown to induce differentiation, cell cycle arrest, apoptosis, autophagy and necrosis in a variety of transformed cell lines. In this review, we mainly discuss how HDACi may elicit a therapeutic response to human cancers through different cell death pathways, in particular, apoptosis and autophagy.
Collapse
Affiliation(s)
- Jing Zhang
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Qing Zhong
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
42
|
Histone deacetylase 2 controls p53 and is a critical factor in tumorigenesis. Biochim Biophys Acta Rev Cancer 2014; 1846:524-38. [PMID: 25072962 DOI: 10.1016/j.bbcan.2014.07.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/05/2014] [Accepted: 07/22/2014] [Indexed: 12/21/2022]
Abstract
Histone deacetylase 2 (HDAC2) regulates biological processes by deacetylation of histones and non-histone proteins. HDAC2 is overexpressed in numerous cancer types, suggesting general cancer-relevant functions of HDAC2. In human tumors the TP53 gene encoding p53 is frequently mutated and wild-type p53 is often disarmed. Molecular pathways inactivating wild-type p53 often remain to be defined and understood. Remarkably, current data link HDAC2 to the regulation of the tumor suppressor p53 by deacetylation and to the maintenance of genomic stability. Here, we summarize recent findings on HDAC2 overexpression in solid and hematopoietic cancers with a focus on mechanisms connecting HDAC2 and p53 in vitro and in vivo. In addition, we present an evidence-based model that integrates molecular pathways and feedback loops by which p53 and further transcription factors govern the expression and the ubiquitin-dependent proteasomal degradation of HDAC2 and of p53 itself. Understanding the interactions between p53 and HDAC2 might aid in the development of new therapeutic approaches against cancer.
Collapse
|
43
|
Barazzuol L, Jeynes JCG, Merchant MJ, Wéra AC, Barry MA, Kirkby KJ, Suzuki M. Radiosensitization of glioblastoma cells using a histone deacetylase inhibitor (SAHA) comparing carbon ions with X-rays. Int J Radiat Biol 2014; 91:90-8. [DOI: 10.3109/09553002.2014.946111] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
44
|
Duncan MT, Shin S, Wu JJ, Mays Z, Weng S, Bagheri N, Miller WM, Shea LD. Dynamic transcription factor activity profiles reveal key regulatory interactions during megakaryocytic and erythroid differentiation. Biotechnol Bioeng 2014; 111:2082-94. [PMID: 24853077 DOI: 10.1002/bit.25262] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 02/23/2014] [Accepted: 03/31/2014] [Indexed: 01/19/2023]
Abstract
The directed differentiation toward erythroid (E) or megakaryocytic (MK) lineages by the MK-E progenitor (MEP) could enhance the ex vivo generation of red blood cells and platelets for therapeutic transfusions. The lineage choice at the MEP bifurcation is controlled in large part by activity within the intracellular signal transduction network, the output of which determines the activity of transcription factors (TFs) and ultimately gene expression. Although many TFs have been implicated, E or MK differentiation is a complex process requiring multiple days, and the dynamics of TF activities during commitment and terminal maturation are relatively unexplored. Herein, we applied a living cell array for the large-scale, dynamic quantification of TF activities during MEP bifurcation. A panel of hematopoietic TFs (GATA-1, GATA-2, SCL/TAL1, FLI-1, NF-E2, PU.1, c-Myb) was characterized during E and MK differentiation of bipotent K562 cells. Dynamic TF activity profiles associated with differentiation towards each lineage were identified, and validated with previous reports. From these activity profiles, we show that GATA-1 is an important hub during early hemin- and PMA-induced differentiation, and reveal several characteristic TF interactions for E and MK differentiation that confirm regulatory mechanisms documented in the literature. Additionally, we highlight several novel TF interactions at various stages of E and MK differentiation. Furthermore, we investigated the mechanism by which nicotinamide (NIC) promoted terminal MK maturation using an MK-committed cell line, CHRF-288-11 (CHRF). Concomitant with its enhancement of ploidy, NIC strongly enhanced the activity of three TFs with known involvement in terminal MK maturation: FLI-1, NF-E2, and p53. Dynamic profiling of TF activity represents a novel tool to complement traditional assays focused on mRNA and protein expression levels to understand progenitor cell differentiation.
Collapse
Affiliation(s)
- Mark T Duncan
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois, 60208
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Gaupel AC, Begley T, Tenniswood M. High throughput screening identifies modulators of histone deacetylase inhibitors. BMC Genomics 2014; 15:528. [PMID: 24968945 PMCID: PMC4089024 DOI: 10.1186/1471-2164-15-528] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 06/18/2014] [Indexed: 12/26/2022] Open
Abstract
Background Previous studies from our laboratory and others have demonstrated that in addition to altering chromatin acetylation and conformation, histone deacetylase inhibitors (HDACi) disrupt the acetylation status of numerous transcription factors and other proteins. A whole genome yeast deletion library screen was used to identify components of the transcriptional apparatus that modulate the sensitivity to the hydroxamic acid-based HDACi, CG-1521. Results Screening 4852 haploid Saccharomyces cerevisiae deletion strains for sensitivity to CG-1521 identifies 407 sensitive and 80 resistant strains. Gene ontology (GO) enrichment analysis shows that strains sensitive to CG-1521 are highly enriched in processes regulating chromatin remodeling and transcription as well as other ontologies, including vacuolar acidification and vesicle-mediated transport. CG-1521-resistant strains include those deficient in the regulation of transcription and tRNA modification. Components of the SAGA histone acetyltransferase (HAT) complex are overrepresented in the sensitive strains, including the catalytic subunit, Gcn5. Cell cycle analysis indicates that both the wild-type and gcn5Δ strains show a G1 delay after CG-1521 treatment, however the gcn5Δ strain displays increased sensitivity to CG-1521-induced cell death compared to the wild-type strain. To test whether the enzymatic activity of Gcn5 is necessary in the response to CG-1521, growth assays with a yeast strain expressing a catalytically inactive variant of the Gcn5 protein were performed and the results show that this strain is less sensitive to CG-1521 than the gcn5Δ strain. Conclusion Genome-wide deletion mutant screening identifies biological processes that affect the sensitivity to the HDAC inhibitor CG-1521, including transcription and chromatin remodeling. This study illuminates the pathways involved in the response to CG-1521 in yeast and provides incentives to understand the mechanisms of HDAC inhibitors in cancer cells. The data presented here demonstrate that components of the SAGA complex are involved in mediating the response to CG-1521. Additional experiments suggest that functions other than the acetyltransferase activity of Gcn5 may be sufficient to attenuate the effects of CG-1521 on cell growth. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-528) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Martin Tenniswood
- Department of Biomedical Sciences, School of Public Health, University at Albany, New York 12222, USA.
| |
Collapse
|
46
|
Dayan C, Hales BF. Effects of ethylene glycol monomethyl ether and its metabolite, 2-methoxyacetic acid, on organogenesis stage mouse limbs in vitro. ACTA ACUST UNITED AC 2014; 101:254-61. [PMID: 24798094 PMCID: PMC4227605 DOI: 10.1002/bdrb.21108] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/11/2014] [Accepted: 03/12/2014] [Indexed: 02/05/2023]
Abstract
Exposure to ethylene glycol monomethyl ether (EGME), a glycol ether compound found in numerous industrial products, or to its active metabolite, 2-methoxyacetic acid (2-MAA), increases the incidence of developmental defects. Using an in vitro limb bud culture system, we tested the hypothesis that the effects of EGME on limb development are mediated by 2-MAA-induced alterations in acetylation programming. Murine gestation day 12 embryonic forelimbs were exposed to 3, 10, or 30 mM EGME or 2-MAA in culture for 6 days to examine effects on limb morphology; limbs were cultured for 1 to 24 hr to monitor effects on the acetylation of histones (H3K9 and H4K12), a nonhistone protein, p53 (p53K379), and markers for cell cycle arrest (p21) and apoptosis (cleaved caspase-3). EGME had little effect on limb morphology and no significant effects on the acetylation of histones or p53 or on biomarkers for cell cycle arrest or apoptosis. In contrast, 2-MAA exposure resulted in a significant concentration-dependent increase in limb abnormalities. 2-MAA induced the hyperacetylation of histones H3K9Ac and H4K12Ac at all concentrations tested (3, 10, and 30 mM). Exposure to 10 or 30 mM 2-MAA significantly increased acetylation of p53 at K379, p21 expression, and caspase-3 cleavage. Thus, 2-MAA, the proximate metabolite of EGME, disrupts limb development in vitro, modifies acetylation programming, and induces biomarkers of cell cycle arrest and apoptosis
Collapse
Affiliation(s)
- Caroline Dayan
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | | |
Collapse
|
47
|
Wilking MJ, Singh C, Nihal M, Zhong W, Ahmad N. SIRT1 deacetylase is overexpressed in human melanoma and its small molecule inhibition imparts anti-proliferative response via p53 activation. Arch Biochem Biophys 2014; 563:94-100. [PMID: 24751483 DOI: 10.1016/j.abb.2014.04.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/03/2014] [Accepted: 04/05/2014] [Indexed: 11/30/2022]
Abstract
Melanoma causes more deaths than any other skin cancer, and its incidence in the US continues to rise. Current medical therapies are insufficient to control this deadly neoplasm, necessitating the development of new target-based approaches. The objective of this study was to determine the role and functional significance of the class III histone deacetylase SIRT1 in melanoma. We have found that SIRT1 is overexpressed in clinical human melanoma tissues and human melanoma cell lines (Sk-Mel-2, WM35, G361, A375, and Hs294T) compared to normal skin and normal melanocytes, respectively. In addition, treatment of melanoma cell lines A375, Hs294T, and G361 with Tenovin-1, a small molecule SIRT1 inhibitor, resulted in a significant decrease in cell growth and cell viability. Further, Tenovin-1 treatment also resulted in a marked decrease in the clonogenic survival of melanoma cells. Further experiments showed that the anti-proliferative response of Tenovin-1 was accompanied by an increase in the protein as well as activity of the tumor suppressor p53. This increase in p53 activity was substantiated by an increase in the protein level of its downstream target p21. Overall, these data suggest that small molecule inhibition of SIRT1 causes anti-proliferative effects in melanoma cells. SIRT1 appears to be acting through the activity of the tumor suppressor p53, which is not mutated in the majority of melanomas. However, future detailed studies are needed to further explore the role and mechanism of SIRT1 in melanoma development and progression and its usefulness in melanoma treatment.
Collapse
Affiliation(s)
- Melissa J Wilking
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, USA
| | - Chandra Singh
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, USA
| | - Minakshi Nihal
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, USA
| | - Weixiong Zhong
- Department of Pathology, University of Wisconsin, Madison, WI 53705, USA
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA.
| |
Collapse
|
48
|
Wu J, Du C, Lv Z, Ding C, Cheng J, Xie H, Zhou L, Zheng S. The up-regulation of histone deacetylase 8 promotes proliferation and inhibits apoptosis in hepatocellular carcinoma. Dig Dis Sci 2013; 58:3545-53. [PMID: 24077923 DOI: 10.1007/s10620-013-2867-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 08/23/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND Histone deacetylase 8 (HDAC8), a member of class I HDACs, has been reported to be involved in transcriptional regulation, cell cycle progression, and developmental events, and several studies have shown that HDAC8 plays a critical role in tumorigenesis. However, the expression level and the potential role of HDAC8 in hepatocellular carcinoma (HCC) remain unclear. AIM The purpose of this study was to investigate protein expression of HDAC8 in HCC tissues and the effects of HDAC8 knockdown on the proliferation and apoptosis of liver cancer cells, and to explore the possible mechanisms. METHODS First, we used quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR), immunohistochemical staining, and western blot to examine the mRNA and protein expression of HDAC8 in HCC cell lines and tissues. Then, we assessed the correlation between clinicopathological parameters and the protein expression of HDAC8. Furthermore, we employed the interfering RNA method to explore the potential role of HDAC8 in HCC progression in vitro. RESULTS Our results showed that expression of HDAC8 was significantly up-regulated both in HCC cell lines and tumor tissues compared to human normal liver cell line LO2 and corresponding non-tumor tissues. Moreover, we found that HDAC8 knockdown could dramatically inhibit HCC cell proliferation and enhance the apoptosis rate in vitro. Western blot revealed that intrinsic apoptotic pathway proteins, including BAX, BAD, and BAK, were elevated after HDAC8 knockdown. The cleavage of caspase-3 and PARP, which are downstream of intrinsic apoptotic pathway, were also enhanced. In addition, suppression of HDAC8 also elevated the expression of p53 and acetylation of p53 at Lys382, whereas the acetylation of p53 at Lys373 did not change. CONCLUSIONS Our study revealed that HDAC8 was overexpressed in HCC. HDAC8 knockdown suppresses tumor growth and enhances apoptosis in HCC via elevating the expression of p53 and acetylation of p53 at Lys382. HDAC8 might serve as a potential therapeutic target in HCC.
Collapse
|
49
|
Ramakrishnan S, Ellis L, Pili R. Histone modifications: implications in renal cell carcinoma. Epigenomics 2013; 5:453-62. [PMID: 23895657 DOI: 10.2217/epi.13.40] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In 2012, an estimated 64,770 men and women were diagnosed with malignancy of the kidney and renal pelvis, of which 13,570 succumbed to their disease. Common genetic aberrations in renal cell carcinomas (RCCs) include loss of function of the VHL gene in clear-cell RCC, overexpression of the c-MET gene in papillary RCC type I, deficiency in the FH gene in papillary RCC type II and loss of heterozygozity of the BHD gene in chromophobe RCC. Recent studies illustrate epigenetic silencing of VHL, as well as alterations in histone modifications and their governing enzymes. The possibility of reversing these epigenetic marks has resulted in efforts to target these changes by utilizing inhibitors of HDACs, DNA methyltransferases and, recently, histone methyltransferases in preclinical and clinical studies. This article focuses on potential therapeutic interventions, and the implications of histone modifications and related enzyme alterations in RCC.
Collapse
Affiliation(s)
- Swathi Ramakrishnan
- Roswell Park Cancer Institute, Department of Cancer Pathology & Prevention, Grace Cancer Drug Center, Buffalo, NY, USA
| | | | | |
Collapse
|
50
|
Lee SY, Huang Z, Kang TH, Soong RS, Knoff J, Axenfeld E, Wang C, Alvarez RD, Chen CS, Hung CF, Wu TC. Histone deacetylase inhibitor AR-42 enhances E7-specific CD8⁺ T cell-mediated antitumor immunity induced by therapeutic HPV DNA vaccination. J Mol Med (Berl) 2013; 91:1221-31. [PMID: 23715898 PMCID: PMC3783646 DOI: 10.1007/s00109-013-1054-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED We have previously created a potent DNA vaccine encoding calreticulin linked to the human papillomavirus (HPV) oncogenic protein E7 (CRT/E7). While treatment with the CRT/E7 DNA vaccine generates significant tumor-specific immune responses in vaccinated mice, the potency with the DNA vaccine could potentially be improved by co-administration of a histone deacetylase inhibitor (HDACi) as HDACi has been shown to increase the expression of MHC class I and II molecules. Thus, we aimed to determine whether co-administration of a novel HDACi, AR-42, with therapeutic HPV DNA vaccines could improve the activation of HPV antigen-specific CD8(+) T cells, resulting in potent therapeutic antitumor effects. To do so, HPV-16 E7-expressing murine TC-1 tumor-bearing mice were treated orally with AR-42 and/or CRT/E7 DNA vaccine via gene gun. Mice were monitored for E7-specific CD8(+) T cell immune responses and antitumor effects. TC-1 tumor-bearing mice treated with AR-42 and CRT/E7 DNA vaccine experienced longer survival, decreased tumor growth, and enhanced E7-specific immune response compared to mice treated with AR-42 or CRT/E7 DNA vaccine alone. Additionally, treatment of TC-1 cells with AR-42 increased the surface expression of MHC class I molecules and increased the susceptibility of tumor cells to the cytotoxicity of E7-specific T cells. This study indicates the ability of AR-42 to significantly enhance the potency of the CRT/E7 DNA vaccine by improving tumor-specific immune responses and antitumor effects. Both AR-42 and CRT/E7 DNA vaccines have been used in independent clinical trials; the current study serves as foundation for future clinical trials combining both treatments in cervical cancer therapy. KEY MESSAGE AR-42, a novel HDAC inhibitor, enhances potency of therapeutic HPV DNA vaccines AR-42 treatment leads to strong E7-specific CD8+ T cell immune responses AR-42 improves tumor-specific immunity and antitumor effects elicited by HPV DNA vaccine AR-42 is more potent than clinically available HDACi in combination with HPV DNA vaccine.
Collapse
Affiliation(s)
- Sung Yong Lee
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Internal Medicine, Korea University Medical Center, Seoul, South Korea
| | - Zhuomin Huang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Gynecology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Tae Heung Kang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Ruey-Shyang Soong
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of General Surgery, Chang Gung Memorial Hospital at Keelung, Taiwan
| | - Jayne Knoff
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Ellen Axenfeld
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Chenguang Wang
- Department of Biostatistics and Bioinformatics, Sidney Kimmel Cancer Comprehensive Cancer Center Johns Hopkins University, Baltimore, MD, USA
| | - Ronald D. Alvarez
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham
| | - Ching-Shih Chen
- Division of Medical Chemistry, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - T.-C. Wu
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|