1
|
Aroonthongsawat P, Manocheewa S, Srisawat C, Punnakitikashem P, Suwanwong Y. Enhancement of the in vitro anti-leukemic effect of the histone deacetylase inhibitor romidepsin using Poly-(D, L-lactide-co-glycolide) nanoparticles as a drug carrier. Eur J Pharm Sci 2025; 207:107043. [PMID: 39952370 DOI: 10.1016/j.ejps.2025.107043] [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: 10/08/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
The goal of this work is to develop a delivery system for histone deacetylase inhibitor (HDACi) romidepsin (ROM) using Poly(D, L-lactide-co-glycolide) as a carrier and evaluate its anti-leukemic effects. Romidepsin-loaded nanoparticles (ROM NPs) required for this purpose were fabricated using a single emulsion-solvent evaporation technique. Their physical characteristics and in vitro drug release profiles were studied, alongside biocompatibility and hemocompatibility assessments. Cell viability assays and Annexin V/Propidium Iodide (PI) staining were conducted to evaluate the anti-leukemic and apoptosis induction efficiency of ROM NPs in vitro. ROM NPs displayed a spherical shape with an average hydrodynamic size of about 149.7 ± 8.4 nm, a PDI of 0.11 ± 0.03, and a zeta potential of -25.27 ± 2.12 mV. The nanoparticles demonstrated a high encapsulation efficiency of ROM (∼93 %) and these nanoparticles effectively entered acute leukemia cells, including U937 and Jurkat. ROM NPs also exhibited a prolonged biphasic release pattern, specifically, the initial burst release phase occurred within the first 24 h, followed by a slower, sustained release. Additionally, they showed no hematological or biological toxicity, indicating their potential use for the delivery of anti-cancer drugs through the circulatory system. In tests on acute leukemia cell lines, ROM NPs showed significantly stronger anti-leukemic effects and induced apoptosis to a greater extent compared to free ROM. In summary, ROM NPs represent a promising therapy option for leukemia according to their enhanced anti-leukemic effects. Further modification of this strategy could be performed to enable target specificity, hence minimizing damage to normal cells.
Collapse
Affiliation(s)
- Pinyadapat Aroonthongsawat
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Siriphan Manocheewa
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chatchawan Srisawat
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Bangkok 10700, Thailand; Siriraj Center of Research Excellence in Theranostic Nanomedicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Primana Punnakitikashem
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Bangkok 10700, Thailand; Siriraj Center of Research Excellence in Theranostic Nanomedicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Yaneenart Suwanwong
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Center of Excellence for Biosensors and Bioengineering (CEBB), Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
2
|
Wang C, Li J, Jiang X, Ma X, Zhen W, Tillman L, Weichselbaum RR, Lin W. Bifunctional Metal-Organic Framework Synergistically Enhances Radiotherapy and Activates STING for Potent Cancer Radio-Immunotherapy. Angew Chem Int Ed Engl 2025; 64:e202417027. [PMID: 39375150 DOI: 10.1002/anie.202417027] [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: 09/04/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/09/2024]
Abstract
The activation of the stimulator of interferon genes (STING) protein by cyclic dinucleotide metabolites plays a critical role in antitumor immunity. However, synthetic STING agonists like 4-(5,6-dimethoxybenzo[b]thiophen-2-yl)-4-oxobutanoic acid (MSA-2) exhibit suboptimal pharmacokinetics and fail to sustain STING activation in tumors for effective antitumor responses. Here, we report the design of MOF/MSA-2, a bifunctional MSA-2 conjugated nanoscale metal-organic framework (MOF) based on Hf6 secondary building units (SBUs) and hexakis(4'-carboxy[1,1'-biphenyl]-4-yl)benzene bridging ligands, for potent cancer radio-immunotherapy. By leveraging the high-Z properties of the Hf6 SBUs, the MOF enhances the therapeutic effect of X-ray radiation and elicits potent immune stimulation in the tumor microenvironment. MOF/MSA-2 further enhances radiotherapeutic effects of X-rays by enabling sustained STING activation and promoting the infiltration and activation of immune cells in the tumors. MOF/MSA-2 plus low-dose X-ray irradiation elicits strong STING activation and potent tumor regression, and when combined with an immune checkpoint inhibitor, effectively suppresses both primary and distant tumors through systemic immune activation.
Collapse
Affiliation(s)
- Chaoyu Wang
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, 5758 S Maryland Ave, Chicago, IL 60637, USA
| | - Jinhong Li
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
| | - Xiaomin Jiang
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, 5758 S Maryland Ave, Chicago, IL 60637, USA
| | - Xin Ma
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
| | - Wenyao Zhen
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, 5758 S Maryland Ave, Chicago, IL 60637, USA
| | - Langston Tillman
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, 5758 S Maryland Ave, Chicago, IL 60637, USA
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, 5758 S Maryland Ave, Chicago, IL 60637, USA
| |
Collapse
|
3
|
Khalid S, Kearney M, McReynolds DE. Can social adversity alter the epigenome, trigger oral disease, and affect future generations? Ir J Med Sci 2024; 193:2597-2606. [PMID: 38740675 PMCID: PMC11450135 DOI: 10.1007/s11845-024-03697-3] [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: 02/23/2024] [Accepted: 04/27/2024] [Indexed: 05/16/2024]
Abstract
The nature versus nurture debate has intrigued scientific circles for decades. Although extensive research has established a clear relationship between genetics and disease development, recent evidence has highlighted the insufficiency of attributing adverse health outcomes to genetic factors alone. In fact, it has been suggested that environmental influences, such as socioeconomic position (SEP), may play a much larger role in the development of disease than previously thought, with extensive research suggesting that low SEP is associated with adverse health conditions. In relation to oral health, a higher prevalence of caries (tooth decay) exists among those of low SEP. Although little is known about the biological mechanisms underlying this relationship, epigenetic modifications resulting from environmental influences have been suggested to play an important role. This review explores the intersection of health inequalities and epigenetics, the role of early-life social adversity and its long-term epigenetic impacts, and how those living within the lower hierarchies of the socioeconomic pyramid are indeed at higher risk of developing diseases, particularly in relation to oral health. A deeper understanding of these mechanisms could lead to the development of targeted interventions for individuals of low SEP to improve oral health or identify those who are at higher risk of developing oral disease.
Collapse
Affiliation(s)
- Sakr Khalid
- Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland
| | - Michaela Kearney
- Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland
| | - David E McReynolds
- Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
4
|
Gao Y, Gao J, Mu G, Zhang Y, Huang F, Zhang W, Ren C, Yang C, Liu J. Selectively enhancing radiosensitivity of cancer cells via in situ enzyme-instructed peptide self-assembly. Acta Pharm Sin B 2020; 10:2374-2383. [PMID: 33354508 PMCID: PMC7745053 DOI: 10.1016/j.apsb.2020.07.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/20/2020] [Accepted: 06/23/2020] [Indexed: 01/20/2023] Open
Abstract
The radiotherapy modulators used in clinic have disadvantages of high toxicity and low selectivity. For the first time, we used the in situ enzyme-instructed self-assembly (EISA) of a peptide derivative (Nap-GDFDFpYSV) to selectively enhance the sensitivity of cancer cells with high alkaline phosphatase (ALP) expression to ionizing radiation (IR). Compared with the in vitro pre-assembled control formed by the same molecule, assemblies formed by in situ EISA in cells greatly sensitized the ALP-high-expressing cancer cells to γ-rays, with a remarkable sensitizer enhancement ratio. Our results indicated that the enhancement was a result of fixing DNA damage, arresting cell cycles and inducing cell apoptosis. Interestingly, in vitro pre-formed assemblies mainly localized in the lysosomes after incubating with cells, while the assemblies formed via in situ EISA scattered in the cell cytosol. The accumulation of these molecules in cells could not be inhibited by endocytosis inhibitors. We believed that this molecule entered cancer cells by diffusion and then in situ self-assembled to form nanofibers under the catalysis of endogenous ALP. This study provides a successful example to utilize intracellular in situ EISA of small molecules to develop selective tumor radiosensitizers. The intracellular in situ enzyme-instructed self-assembly (in situ EISA) was firstly used for selective cancer radiosensitization. Compared with the in vitro pre-assembled control formed by the same molecule, assemblies formed by in-situ EISA in cells greatly sensitized the ALP-high-expressing cancer cells to γ-rays. This work provides a successful example to utilize intracellular in situ EISA of small molecules to develop selective tumor radiosensitizers.
Collapse
Affiliation(s)
- Yang Gao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Jie Gao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Ganen Mu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yumin Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Fan Huang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Wenxue Zhang
- Radiation Oncology Department, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chunhua Ren
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
- Corresponding authors.
| | - Cuihong Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
- Corresponding authors.
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
- Corresponding authors.
| |
Collapse
|
5
|
Chen CY, Fang JY, Chen CC, Chuang WY, Leu YL, Ueng SH, Wei LS, Cheng SF, Hsueh C, Wang TH. 2-O-Methylmagnolol, a Magnolol Derivative, Suppresses Hepatocellular Carcinoma Progression via Inhibiting Class I Histone Deacetylase Expression. Front Oncol 2020; 10:1319. [PMID: 32850418 PMCID: PMC7431949 DOI: 10.3389/fonc.2020.01319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/24/2020] [Indexed: 12/24/2022] Open
Abstract
Magnolia officinalis is widely used in Southeast Asian countries for the treatment of fever, headache, diarrhea, and stroke. Magnolol is a phenolic compound extracted from M. officinalis, with proven antibacterial, antioxidant, anti-inflammatory, and anticancer activities. In this study, we modified magnolol to synthesize a methoxylated derivative, 2-O-methylmagnolol (MM1), and investigated the use of MM1, and magnolol in the treatment of liver cancer. We found that both magnolol and MM1 exhibited inhibitory effects on the growth, migration, and invasion of hepatocellular carcinoma (HCC) cell lines and halted the cell cycle at the G1 phase. MM1 also demonstrated a substantially better tumor-suppressive effect than magnolol. Further analysis suggested that by inhibiting class I histone deacetylase expression in HCC cell lines, magnolol and MM1 induced p21 expression and p53 activation, thereby causing cell cycle arrest and inhibiting HCC cell growth, migration, and invasion. Subsequently, we verified the significant tumor-suppressive effects of magnolol and MM1 in an animal model. Collectively, these findings demonstrate the anti-HCC activities of magnolol and MM1 and their potential for clinical use.
Collapse
Affiliation(s)
- Chi-Yuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| | - Jia-You Fang
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan City, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Chin-Chuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Taoyuan City, Taiwan
| | - Wen-Yu Chuang
- Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan City, Taiwan
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan City, Taiwan.,Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan.,Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Shir-Hwa Ueng
- Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan City, Taiwan
| | - Li-Shan Wei
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Shu-Fang Cheng
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Taoyuan City, Taiwan
| | - Chuen Hsueh
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan City, Taiwan
| | - Tong-Hong Wang
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan.,Department of Hepato-Gastroenterology, Liver Research Center, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| |
Collapse
|
6
|
Reda M, Bagley AF, Zaidan HY, Yantasee W. Augmenting the therapeutic window of radiotherapy: A perspective on molecularly targeted therapies and nanomaterials. Radiother Oncol 2020; 150:225-235. [PMID: 32598976 DOI: 10.1016/j.radonc.2020.06.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/25/2022]
Abstract
Radiation therapy is a cornerstone of modern cancer therapy alongside surgery, chemotherapy, and immunotherapy, with over half of all cancer patients receiving radiation therapy as part of their treatment regimen. Development of novel radiation sensitizers that can improve the therapeutic window of radiation therapy are sought after, particularly for tumors at an elevated risk of local and regional recurrence such as locally-advanced lung, head and neck, and gastrointestinal tumors. This review discusses clinical strategies to enhance radiotherapy efficacy and decrease toxicity, hence, increasing the overall therapeutic window. A focus is given to the molecular targets that have been identified and their associated mechanisms of action in enhancing radiotherapy. Examples include cell survival and proliferation signaling such as the EGFR and PI3K/AKT/mTOR pathways, DNA repair genes including PARP and ATM/ATR, angiogenic growth factors, epigenetic regulators, and immune checkpoint proteins. By manipulating various mechanisms of tumor resistance to ionizing radiation (IR), targeted therapies hold significant value to increase the therapeutic window of radiotherapy. Further, the use of novel nanoparticles to enhance radiotherapy is also reviewed, including nanoparticle delivery of chemotherapies, metallic (high-Z) nanoparticles, and nanoparticle delivery of targeted therapies - all of which may improve the therapeutic window of radiotherapy by enhancing the tumor response to IR or reducing normal tissue toxicity.
Collapse
Affiliation(s)
- Moataz Reda
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, United States; PDX Pharmaceuticals, Portland, OR 97239, United States
| | - Alexander F Bagley
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | | | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, United States; PDX Pharmaceuticals, Portland, OR 97239, United States.
| |
Collapse
|
7
|
Chen CY, Chen CC, Chuang WY, Leu YL, Ueng SH, Hsueh C, Yeh CT, Wang TH. Hydroxygenkwanin Inhibits Class I HDAC Expression and Synergistically Enhances the Antitumor Activity of Sorafenib in Liver Cancer Cells. Front Oncol 2020; 10:216. [PMID: 32158695 PMCID: PMC7052045 DOI: 10.3389/fonc.2020.00216] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/07/2020] [Indexed: 12/13/2022] Open
Abstract
Abnormal histone deacetylase (HDAC) expression is closely related to cancer development and progression. Many HDAC inhibitors have been widely used in cancer treatment; however, severe side effects often limit their clinical application. In this study, we attempted to identify natural compounds with HDAC inhibitory activity and low physiological toxicity and explored their feasibility and mechanisms of action in liver cancer treatment. A yeast screening system was used to identify natural compounds with HDAC inhibitory activity. Further, western blotting was used to verify inhibitory effects on HDAC in human liver cancer cell lines. Cell functional analysis was used to explore the effects and mechanisms and the in vitro results were verified in BALB/c nude mice. We found that hydroxygenkwanin (HGK), an extract from Daphne genkwa, inhibited class I HDAC expression, and thereby induced expression of tumor suppressor p21 and promoted acetylation and activation of p53 and p65. This resulted in the inhibition of growth, migration, and invasion of liver cancer cells and promoted cell apoptosis. Animal models revealed that HGK inhibited tumor growth in a synergistic manner with sorafenib. HGK inhibited class I HDAC expression and had low physiological toxicity. It has great potential as an adjuvant for liver cancer treatment and may be used in combination with anticancer drugs like sorafenib to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Chi-Yuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Chin-Chuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Yu Chuang
- Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan.,Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.,Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shir-Hwa Ueng
- Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Chuen Hsueh
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Chau-Ting Yeh
- Department of Hepato-Gastroenterology, Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Tong-Hong Wang
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Hepato-Gastroenterology, Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
8
|
Jang B, Yang IH, Cho NP, Jin B, Lee W, Jung YC, Hong SD, Shin JA, Cho SD. Down-regulation and nuclear localization of survivin by sodium butyrate induces caspase-dependent apoptosis in human oral mucoepidermoid carcinoma. Oral Oncol 2019; 88:160-167. [PMID: 30616788 DOI: 10.1016/j.oraloncology.2018.11.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/20/2018] [Accepted: 11/29/2018] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Sodium butyrate (NaBu) is a histone deacetylase inhibitor that possesses an apoptotic ability. However, the molecular mechanism by which NaBu induces apoptosis in human oral mucoepidermoid carcinoma (MEC), a type of salivary gland tumor, remains unclear. MATERIALS AND METHODS The anticancer effects of NaBu and its related molecular mechanisms were determined by trypan blue exclusion assay, 4'-6-diamidino-2-phenylindole staining, live/dead assay, human apoptosis array, RT-PCR, western blotting, immunocytochemistry, preparation of nuclear fractions, and nude mice tumor xenograft. RESULTS In this study, we found that NaBu inhibited growth and induced apoptosis in the human oral MEC cell lines MC3 and YD15 with acetylation of histone proteins H2A and H3. NaBu apparently down-regulated survivin protein, as evidenced by the results of the human apoptosis antibody array, and modulated it at the post-translational process. Interestingly, NaBu caused nuclear translocation of survivin protein in both cell lines. NaBu also resulted in decreased expression levels of Bcl-xL mRNA and protein, leading to induction of caspase-dependent apoptosis in human oral MEC cell lines. In addition, NaBu administration inhibited tumor growth in vivo at a dosage of 500 mg/kg/day, but it did not cause any hepatic or renal toxicity. CONCLUSION This study provides new insights into the molecular mechanism of apoptotic actions by NaBu in human oral MEC and the basis of its clinical application for the treatment of human oral MEC.
Collapse
Affiliation(s)
- Boonsil Jang
- Department of Dental Hygiene, Sorabol College, Gyeongju 38063, Republic of Korea
| | - In-Hyoung Yang
- Department of Oral Pathology, School of Dentistry, Institute of Oral Bioscience and Biodegradable Material, Chonbuk National University, Jeonju 54896, Republic of Korea
| | - Nam-Pyo Cho
- Department of Oral Pathology, School of Dentistry, Institute of Oral Bioscience and Biodegradable Material, Chonbuk National University, Jeonju 54896, Republic of Korea
| | - Bohwan Jin
- Laboratory Animal Center, CHA University, CHA Biocomplex, Sampyeong-dong, Seongnam 13488, Republic of Korea
| | - WonWoo Lee
- Laboratory Animal Center, CHA University, CHA Biocomplex, Sampyeong-dong, Seongnam 13488, Republic of Korea
| | - Yun Chan Jung
- Chaon, 301-3, 240, Pangyoyeok-ro, Bundang-gu, Seongnam, Republic of Korea
| | - Seong Doo Hong
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Ji-Ae Shin
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea.
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea.
| |
Collapse
|
9
|
Attia SM, Al-Hamamah MA, Alotaibi MR, Harisa GI, Attia MM, Ahmad SF, Ansari MA, Nadeem A, Bakheet SA. Investigation of belinostat-induced genomic instability by molecular cytogenetic analysis and pathway-focused gene expression profiling. Toxicol Appl Pharmacol 2018; 350:43-51. [PMID: 29733868 DOI: 10.1016/j.taap.2018.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/16/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022]
Abstract
Histone deacetylases (HDACs), which regulate transcription and specific functions such as tumor suppression by p53, are frequently altered in tumors and have a contentious role in carcinogenesis. HDAC inhibitors, which have a long history of use in psychiatry and neurology, have recently been tested as possible treatments for tumors. Belinostat received regulatory approval in the USA on July 3, 2014, for use against peripheral T-cell lymphoma. However, the unavailability of information on belinostat genotoxicity in normal cells and the molecular mechanisms involved in the genetic instability after exposure to belinostat encouraged us to conduct this study. Our data showed that the exposure of mice to belinostat at the recommended human doses induced chromosome breakage, whole-chromosome lagging, and oxidative DNA damage in bone marrow cells in a dose-dependent manner. The expression levels of 84 genes involved in the DNA damage signaling pathway were evaluated by using an RT2 Profiler PCR array. Belinostat exposure altered the expression of 25 genes, with statistically significant changes observed in 17 genes. The array results were supported by RT-PCR and western blotting experiments. Collectively, our results showed that belinostat exposure caused oxidative DNA damage and downregulated the expression of genes involved in DNA damage repair, which may be responsible for belinostat-induced genomic instability. Thus, the clinical usage of this drug should be weighed against the hazards of carcinogenesis, and the observed genotoxicity profile of belinostat may support further development of efficient HDAC inhibitors with weaker genotoxicity.
Collapse
Affiliation(s)
- S M Attia
- Pharmacology & Toxicology Department, Faculty of Pharmacy, King Saud University, P.O. 2457, Riyadh 11451, Saudi Arabia; Pharmacology & Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt.
| | - M A Al-Hamamah
- Pharmacology & Toxicology Department, Faculty of Pharmacy, King Saud University, P.O. 2457, Riyadh 11451, Saudi Arabia
| | - M R Alotaibi
- Pharmacology & Toxicology Department, Faculty of Pharmacy, King Saud University, P.O. 2457, Riyadh 11451, Saudi Arabia
| | - G I Harisa
- Kayyali Chair for Pharmaceutical Industry, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - M M Attia
- Plant Protection Department, College of Agriculture, Damanhour University, Damanhour, Egypt
| | - S F Ahmad
- Pharmacology & Toxicology Department, Faculty of Pharmacy, King Saud University, P.O. 2457, Riyadh 11451, Saudi Arabia
| | - M A Ansari
- Pharmacology & Toxicology Department, Faculty of Pharmacy, King Saud University, P.O. 2457, Riyadh 11451, Saudi Arabia
| | - A Nadeem
- Pharmacology & Toxicology Department, Faculty of Pharmacy, King Saud University, P.O. 2457, Riyadh 11451, Saudi Arabia
| | - S A Bakheet
- Pharmacology & Toxicology Department, Faculty of Pharmacy, King Saud University, P.O. 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
10
|
Seshacharyulu P, Baine MJ, Souchek JJ, Menning M, Kaur S, Yan Y, Ouellette MM, Jain M, Lin C, Batra SK. Biological determinants of radioresistance and their remediation in pancreatic cancer. Biochim Biophys Acta Rev Cancer 2017; 1868:69-92. [PMID: 28249796 PMCID: PMC5548591 DOI: 10.1016/j.bbcan.2017.02.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/16/2017] [Accepted: 02/17/2017] [Indexed: 12/17/2022]
Abstract
Despite recent advances in radiotherapy, a majority of patients diagnosed with pancreatic cancer (PC) do not achieve objective responses due to the existence of intrinsic and acquired radioresistance. Identification of molecular mechanisms that compromise the efficacy of radiation therapy and targeting these pathways is paramount for improving radiation response in PC patients. In this review, we have summarized molecular mechanisms associated with the radio-resistant phenotype of PC. Briefly, we discuss the reversible and irreversible biological consequences of radiotherapy, such as DNA damage and DNA repair, mechanisms of cancer cell survival and radiation-induced apoptosis following radiotherapy. We further describe various small molecule inhibitors and molecular targeting agents currently being tested in preclinical and clinical studies as potential radiosensitizers for PC. Notably, we draw attention towards the confounding effects of cancer stem cells, immune system, and the tumor microenvironment in the context of PC radioresistance and radiosensitization. Finally, we discuss the need for examining selective radioprotectors in light of the emerging evidence on radiation toxicity to non-target tissue associated with PC radiotherapy.
Collapse
Affiliation(s)
- Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Michael J Baine
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Joshua J Souchek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Melanie Menning
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ying Yan
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Michel M. Ouellette
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Chi Lin
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
11
|
Nickoloff JA, Jones D, Lee SH, Williamson EA, Hromas R. Drugging the Cancers Addicted to DNA Repair. J Natl Cancer Inst 2017; 109:3832892. [PMID: 28521333 PMCID: PMC5436301 DOI: 10.1093/jnci/djx059] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 03/10/2017] [Indexed: 12/29/2022] Open
Abstract
Defects in DNA repair can result in oncogenic genomic instability. Cancers occurring from DNA repair defects were once thought to be limited to rare inherited mutations (such as BRCA1 or 2). It now appears that a clinically significant fraction of cancers have acquired DNA repair defects. DNA repair pathways operate in related networks, and cancers arising from loss of one DNA repair component typically become addicted to other repair pathways to survive and proliferate. Drug inhibition of the rescue repair pathway prevents the repair-deficient cancer cell from replicating, causing apoptosis (termed synthetic lethality). However, the selective pressure of inhibiting the rescue repair pathway can generate further mutations that confer resistance to the synthetic lethal drugs. Many such drugs currently in clinical use inhibit PARP1, a repair component to which cancers arising from inherited BRCA1 or 2 mutations become addicted. It is now clear that drugs inducing synthetic lethality may also be therapeutic in cancers with acquired DNA repair defects, which would markedly broaden their applicability beyond treatment of cancers with inherited DNA repair defects. Here we review how each DNA repair pathway can be attacked therapeutically and evaluate DNA repair components as potential drug targets to induce synthetic lethality. Clinical use of drugs targeting DNA repair will markedly increase when functional and genetic loss of repair components are consistently identified. In addition, future therapies will exploit artificial synthetic lethality, where complementary DNA repair pathways are targeted simultaneously in cancers without DNA repair defects.
Collapse
Affiliation(s)
- Jac A Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Dennie Jones
- Department of Medicine and the Cancer Center, University of Florida Health, Gainesville, FL, USA
| | - Suk-Hee Lee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Elizabeth A Williamson
- Department of Medicine and the Cancer Center, University of Florida Health, Gainesville, FL, USA
| | - Robert Hromas
- Department of Medicine and the Cancer Center, University of Florida Health, Gainesville, FL, USA
| |
Collapse
|
12
|
Nam JH, Cho H, Kang H, Lee J, Jung M, Chang Y, Kim K, Hoe H. A Mercaptoacetamide‐Based Class II Histone Deacetylase Inhibitor Suppresses Cell Migration and Invasion in Monomorphic Malignant Human Glioma Cells by Inhibiting FAK/STAT3 Signaling. J Cell Biochem 2017; 118:4672-4685. [DOI: 10.1002/jcb.26133] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/11/2017] [Indexed: 01/02/2023]
Affiliation(s)
- Jin Han Nam
- Department of Neural Development and DiseaseKorea Brain Research Institute (KBRI)61, Cheomdan‐ro, Dong‐guDaegu41068Korea
| | - Hyun‐Ji Cho
- Department of Neural Development and DiseaseKorea Brain Research Institute (KBRI)61, Cheomdan‐ro, Dong‐guDaegu41068Korea
| | - Hyejin Kang
- Department of Neural Development and DiseaseKorea Brain Research Institute (KBRI)61, Cheomdan‐ro, Dong‐guDaegu41068Korea
| | - Ju‐Young Lee
- Department of Neural Development and DiseaseKorea Brain Research Institute (KBRI)61, Cheomdan‐ro, Dong‐guDaegu41068Korea
| | - Mira Jung
- Department of Radiation MedicineLombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDistrict Of Columbia, DC20057‐1464
| | - Young‐Chae Chang
- Research Institute of Biomedical Engineering and Department of MedicineCatholic University of Daegu School of MedicineDaegu42472Korea
| | - Keetae Kim
- Department of New BiologyDGISTDaegu42988Korea
| | - Hyang‐Sook Hoe
- Department of Neural Development and DiseaseKorea Brain Research Institute (KBRI)61, Cheomdan‐ro, Dong‐guDaegu41068Korea
| |
Collapse
|
13
|
Song JM, Sung YM, Nam JH, Yoon H, Chung A, Moffat E, Jung M, Pak DTS, Kim J, Hoe HS. A Mercaptoacetamide-Based Class II Histone Deacetylase Inhibitor Increases Dendritic Spine Density via RasGRF1/ERK Pathway. J Alzheimers Dis 2016; 51:591-604. [PMID: 26890742 DOI: 10.3233/jad-150717] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The accumulation of amyloid-β (Aβ) leads to the loss of dendritic spines and synapses, which is hypothesized to cause cognitive impairments in Alzheimer's disease (AD) patients. In our previous study, we demonstrated that a novel mercaptoacetamide-based class II histone deacetylase inhibitor (HDACI), known as W2, decreased Aβ levels and improved learning and memory in mice. However, the underlying mechanism of this effect is unknown. OBJECTIVE Because dendritic spine formation is associated with cognitive performance, here we investigated whether HDACI W2 regulates dendritic spine density and its molecular mechanism of action. METHODS To examine the effect of HDACI W2 on dendritic spine density, we conducted morphological analysis of dendritic spines using GFP transfection and Golgi staining. In addition, to determine the molecular mechanism of W2 effects on spines, we measured the levels of mRNAs and proteins involved in the Ras signaling pathway using quantitative real-time PCR, immunocytochemistry, and western analysis. RESULTS We found that HDACI W2 altered dendritic spine density and morphology in vitro and in vivo. Additionally, W2 increased the mRNA or protein levels of Ras GRF1 and phospho-ERK. Moreover, knockdown of RasGRF1 and inhibition of ERK activity prevented the W2-mediated spinogenesis in primary hippocampal neurons. CONCLUSION Our Class II-selective HDACI W2 promotes the formation and growth of dendritic spines in a RasGRF1 and ERK dependent manner in primary hippocampal neurons.
Collapse
Affiliation(s)
- Jung Min Song
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - You Me Sung
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Jin Han Nam
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, Korea
| | - Hyejin Yoon
- Department of Neuroscience, Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Andrew Chung
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Emily Moffat
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Mira Jung
- Department of Radiation Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Daniel T S Pak
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC, USA
| | - Jungsu Kim
- Department of Neuroscience, Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Hyang-Sook Hoe
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA.,Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, Korea
| |
Collapse
|
14
|
Schaue D, McBride WH. Opportunities and challenges of radiotherapy for treating cancer. Nat Rev Clin Oncol 2015; 12:527-40. [PMID: 26122185 DOI: 10.1038/nrclinonc.2015.120] [Citation(s) in RCA: 492] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The past 20 years have seen dramatic changes in the delivery of radiation therapy, but the impact of radiobiology on the clinic has been far less substantial. A major consideration in the use of radiotherapy has been on how best to exploit differences between the tumour and host tissue characteristics, which in the past has been achieved empirically by radiation-dose fractionation. New advances are uncovering some of the mechanistic processes that underlie this success story. In this Review, we focus on how these processes might be targeted to improve the outcome of radiotherapy at the individual patient level. This approach would seem a more productive avenue of treatment than simply trying to increase the radiation dose delivered to the tumour.
Collapse
Affiliation(s)
- Dörthe Schaue
- Department of Radiation Oncology, Room B3-109, Center for Health Sciences, Westwood, University of California, Los Angeles (UCLA), Los Angeles, CA 90095-1714, USA
| | - William H McBride
- Department of Radiation Oncology, Room B3-109, Center for Health Sciences, Westwood, University of California, Los Angeles (UCLA), Los Angeles, CA 90095-1714, USA
| |
Collapse
|
15
|
Bioprotective Carnitinoids: Lipoic Acid, Butyrate, and Mitochondria-Targeting to Treat Radiation Injury: Mitochondrial Drugs Come of Age. Drug Dev Res 2015; 76:167-75. [DOI: 10.1002/ddr.21258] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 05/28/2015] [Indexed: 12/16/2022]
|
16
|
Coiffier B, Federico M, Caballero D, Dearden C, Morschhauser F, Jäger U, Trümper L, Zucca E, Gomes da Silva M, Pettengell R, Weidmann E, d'Amore F, Tilly H, Zinzani PL. Therapeutic options in relapsed or refractory peripheral T-cell lymphoma. Cancer Treat Rev 2014; 40:1080-8. [PMID: 25199959 DOI: 10.1016/j.ctrv.2014.08.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 07/29/2014] [Accepted: 08/14/2014] [Indexed: 11/28/2022]
Abstract
Peripheral T-cell lymphoma (PTCL) represents a relatively rare group of heterogeneous non-Hodgkin lymphomas with a very poor prognosis. Current therapies, based on historical regimens for aggressive B-cell lymphomas, have resulted in insufficient patient outcomes. The majority of patients relapse rapidly, and current 5-year overall survival rates are only 10-30%. It is evident that new approaches to treat patients with PTCL are required. In recent years, prospective studies in PTCL have been initiated, mainly in patients with relapsed/refractory disease. In some of these, selected histologic subtypes have been evaluated in detail. As a consequence, numerous new therapies have been developed and shown activity in PTCL, including: agents targeting the immune system (e.g. brentuximab vedotin, alemtuzumab, lenalidomide); histone deacetylase inhibitors (romidepsin, belinostat); antifolates (pralatrexate); fusion proteins (denileukin diftitox); nucleoside analogs (pentostatin, gemcitabine); and other agents (e.g. alisertib, plitidepsin, bendamustine, bortezomib). A variety of interesting novel combinations is also emerging. It is hoped that these innovative approaches, coupled with a greater understanding of the clinicopathologic features, pathogenesis, molecular biology, and natural history of PTCL will advance the field and improve outcomes in this challenging group of diseases. This review summarizes the currently available clinical evidence on the various approaches to treating relapsed/refractory PTCL, including the role of stem cell transplantation, with an emphasis on potential new drug therapies.
Collapse
Affiliation(s)
| | - Massimo Federico
- Dipartimento di Medicina di Laboratorio, Clinica e di Sanità Pubblica, Università degli studi di Modena e Reggio Emilia, Policlinico, Via del Pozzo, 71, 41124 Modena, Italy.
| | - Dolores Caballero
- Instituto Biosanitario de Salamanca, Paseo de San Vicente 58-182, 37007 Salamanca, Spain.
| | - Claire Dearden
- Department of Haemato-Oncology, Royal Marsden Hospital, Downs Road, SM2 5PT Sutton, UK.
| | - Franck Morschhauser
- Department of Hematology, University Hospital of Lille, F-59037 Lille, France.
| | - Ulrich Jäger
- Medical University of Vienna, Department of Medicine I, Division of Hematology and Hemostaseology, Comprehensive Cancer Center, Waehringer Guertel 18-20, 1090 Vienna, Austria.
| | - Lorenz Trümper
- UniversitätsKrebszentrum (G-CCC), Georg August University, 37099 Göttingen, Germany.
| | - Emanuele Zucca
- Oncology Institute of Southern Switzerland (IOSI), Ospedale San Giovanni, 6500 Bellinzona, Switzerland.
| | - Maria Gomes da Silva
- Instituto Português de Oncologia de Lisboa de Francisco Gentil, R. Prof. Lima Basto, 1099-023 Lisbon, Portugal.
| | - Ruth Pettengell
- St George's University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Eckhart Weidmann
- Klinik für Onkologie und Hämatologie am Krankenhaus Nordwest GmbH, Steinbacher Hohl 2-26, D-60488 Frankfurt, Germany.
| | - Francesco d'Amore
- Department Hematology, Aarhus University Hospital, Tage Hansens Gade 2, DK-8000 Aarhus C, Denmark.
| | - Hervé Tilly
- Department of Hematology, Centre Henri-Becquerel, UMR918, Université de Rouen, Rue d'Amiens, 76038 Rouen Cedex 1, France.
| | - Pier Luigi Zinzani
- Institute of Hematology "Seràgnoli", University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| |
Collapse
|
17
|
MCM-2 is a therapeutic target of Trichostatin A in colon cancer cells. Toxicol Lett 2013; 221:23-30. [DOI: 10.1016/j.toxlet.2013.05.643] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/02/2013] [Accepted: 05/30/2013] [Indexed: 12/17/2022]
|
18
|
Carrier F. Chromatin Modulation by Histone Deacetylase Inhibitors: Impact on Cellular Sensitivity to Ionizing Radiation. MOLECULAR AND CELLULAR PHARMACOLOGY 2013; 5:51-59. [PMID: 24648865 PMCID: PMC3955893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
It is well established that cells are more sensitive to ionizing radiation during the G2/M phase of the cell cycle when their chromatin is highly compacted. However, highly compacted chromatin is less susceptible to DNA Double Strand Breaks (DSBs) than relaxed chromatin. Therefore, it is now becoming apparent that it is the cell capacity to repair its damaged DNA and refold its chromatin into its original compacted status that primarily affects the overall cellular sensitivity to ionizing radiation. The Histone Deacetylase Inhibitors (HDACIs) are a new class of anticancer agents that relax chromatin structure by increasing the levels of histone acetylation. The effect of HDACIs on normal and cancer cells sensitivity to ionizing radiation differs. Reports have indicated that HDACIs can protect normal cells while simultaneously sensitize cancer cells to ionizing radiation. This difference may stem from the individual characteristic of the normal and cancer cells chromatin structure. This review discusses this possibility and addresses the role of HDACIs in radiation therapy.
Collapse
Affiliation(s)
- France Carrier
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland, Baltimore, School of Medicine, Department of Radiation Oncology, Baltimore, Maryland
| |
Collapse
|
19
|
Friedl AA, Mazurek B, Seiler DM. Radiation-induced alterations in histone modification patterns and their potential impact on short-term radiation effects. Front Oncol 2012; 2:117. [PMID: 23050241 PMCID: PMC3445916 DOI: 10.3389/fonc.2012.00117] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 08/28/2012] [Indexed: 12/12/2022] Open
Abstract
Detection and repair of radiation-induced DNA damage occur in the context of chromatin. An intricate network of mechanisms defines chromatin structure, including DNA methylation, incorporation of histone variants, histone modifications, and chromatin remodeling. In the last years it became clear that the cellular response to radiation-induced DNA damage involves all of these mechanisms. Here we focus on the current knowledge on radiation-induced alterations in post-translational histone modification patterns and their effect on the chromatin accessibility, transcriptional regulation and chromosomal stability.
Collapse
Affiliation(s)
- Anna A Friedl
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Germany
| | | | | |
Collapse
|