151
|
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
|
152
|
Kalyaanamoorthy S, Chen YPP. A steered molecular dynamics mediated hit discovery for histone deacetylases. Phys Chem Chem Phys 2014; 16:3777-91. [PMID: 24429775 DOI: 10.1039/c3cp53511h] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The inhibitors of class I histone deacetylases (HDACIs) have gained significant interest in cancer therapeutics. Virtual high throughput screening (vHTS) is one of the popular approaches used in the identification of novel scaffolds of HDACIs. However, an accurate description of ligand-protein flexibilities in the vHTS remains challenging. In this work, we implement an integrated approach, which combines the vHTS with the 'state-of-the-art' steered molecular dynamics (SMD). This approach serves as an efficient tool to identify potential hits and characterize their binding potencies against the class I HDACs in a flexible solvent environment. A hybrid pharmacophore-based and structure-based vHTS method identifies the hits with more favourable physico-chemical features against the class I HDACs. Our pharmacophore-based screening enhanced the quality of the vHTS outcomes. Further, the molecular interactions between the hits and the HDACs are investigated using the SMD-driven force profiles, which in turn resulted in filtering the hits with higher binding potencies against the HDACs. Our results, therefore, reveal that vHTS and SMD can be a complementary and effective analytical tool for accelerating the hit identification phase in structure-based drug design.
Collapse
Affiliation(s)
- Subha Kalyaanamoorthy
- Faculty of Science, Technology and Engineering, La Trobe University, Melbourne, Victoria 3086, Australia.
| | | |
Collapse
|
153
|
Tasoulas J, Giaginis C, Patsouris E, Manolis E, Theocharis S. Histone deacetylase inhibitors in oral squamous cell carcinoma treatment. Expert Opin Investig Drugs 2014; 24:69-78. [PMID: 25216628 DOI: 10.1517/13543784.2014.952368] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Introduction: The involvement of the histone deacetylases (HDACs) family in tumor development and progression is well demonstrated. HDAC inhibitors (HDACis) constitute a novel, heterogeneous family of highly selective anticancer agents that inhibit HDACs and present significant antitumor activity in several human malignancies, including oral squamous cell carcinoma (OSCC). Areas covered: This review summarizes the current research on the anticancer activity of HDACis against OSCC. The review also presents the molecular mechanisms of HDACis action and the existing studies evaluating their utilization in combined therapies of OSCC. Expert opinion: The currently available data support evidence that HDACis may provide new therapeutic options against OSCC, decreasing treatment side effects and allowing a more conservative therapeutic approach. Future research should be focused on in vivo and clinical evaluation of their utilization as combined therapies or monotherapies. Before HDACis can be brought into clinical practice as treatment options for OSCC, further evaluation is needed to determine their optimal dosage, the appropriate duration of treatment and whether they should be used in combination or as stand-alone therapeutics.
Collapse
Affiliation(s)
- Jason Tasoulas
- National and Kapodistrian University of Athens, Medical School, First Department of Pathology , Athens , Greece
| | | | | | | | | |
Collapse
|
154
|
Foss F, Coiffier B, Horwitz S, Pro B, Prince HM, Sokol L, Greenwood M, Lerner A, Caballero D, Baran E, Kim E, Nichols J, Balser B, Wolfson J, Whittaker S. Tolerability to romidepsin in patients with relapsed/refractory T-cell lymphoma. Biomark Res 2014; 2:16. [PMID: 25279222 PMCID: PMC4181623 DOI: 10.1186/2050-7771-2-16] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 06/27/2014] [Indexed: 11/11/2022] Open
Abstract
Background Histone deacetylase inhibitor romidepsin has demonstrated durable clinical responses and tolerability in patients with relapsed/refractory peripheral and cutaneous T-cell lymphoma (PTCL, CTCL). Selection of novel drug therapies for patients with relapsed/refractory aggressive lymphoma requires not only considerations regarding efficacy but also careful evaluation of toxicities as well as overall clinical benefit. The purpose of this analysis was to examine common adverse events (AEs) reported in pivotal trials of romidepsin in relapsed/refractory PTCL or CTCL and to more clearly define the overall AE profile in these populations. Methods Patients with relapsed/refractory PTCL or CTCL were treated with romidepsin at 14 mg/m2 as a 4-hour intravenous infusion on days 1, 8, and 15 of 28-day cycles for up to 6 cycles; patients with at least stable disease could extend therapy until progressive disease or another withdrawal criterion was met. All enrolled patients who received ≥ 1 dose of romidepsin were included in the AE analyses. Results Overall, safety profiles of common AEs were similar, although patients with relapsed/refractory PTCL had more frequent hematologic toxicities and grade ≥ 3 infections. In both patient populations, the greatest incidence of grade ≥ 3 AEs and the majority of discontinuations due to AEs occurred during cycles 1–2. Early discontinuations were primarily related to infection, thrombocytopenia, or electrocardiogram abnormalities, confirming the need to closely monitor patients with poor bone marrow reserve or other comorbidities. Despite this, 28% of patients with relapsed/refractory PTCL and 36% of patients with relapsed/refractory CTCL continued on romidepsin treatment for ≥ 6 cycles. Conclusions This study demonstrates that patients with relapsed/refractory PTCL or CTCL have similar AE profiles with romidepsin treatment, although patients with PTCL experienced more frequent and more severe hematologic toxicities and more frequent grade ≥ 3 infections. The greatest incidence of grade ≥ 3 AEs and the majority of discontinuations due to AEs occurred during treatment cycles 1–2. Extended dosing of romidepsin can be tolerated in responding patients. Trial registration NCT00426764,NCT00106431
Collapse
Affiliation(s)
- Francine Foss
- Yale Cancer Center, PO Box 208028, 333 Cedar Street, TMP 3, New Haven, CT 06520-8028, USA
| | | | - Steven Horwitz
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Barbara Pro
- Thomas Jefferson University, Philadelphia, PA, USA
| | - H Miles Prince
- Peter MacCallum Cancer Centre and University of Melbourne, East Melbourne, Victoria, Australia
| | | | | | | | | | | | - Ellen Kim
- University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - Sean Whittaker
- Guy's and St Thomas' Hospital, London, England, United Kingdom
| |
Collapse
|
155
|
Hrzenjak A, Dieber-Rotheneder M, Moinfar F, Petru E, Zatloukal K. Molecular mechanisms of endometrial stromal sarcoma and undifferentiated endometrial sarcoma as premises for new therapeutic strategies. Cancer Lett 2014; 354:21-7. [PMID: 25128649 DOI: 10.1016/j.canlet.2014.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 08/11/2014] [Accepted: 08/11/2014] [Indexed: 11/28/2022]
Abstract
Endometrial stromal sarcoma (ESS) and undifferentiated endometrial sarcoma (UES) are very rare gynecologic malignancies. Due to the rarity and heterogeneity of these tumors, little is known about their epidemiology, pathogenesis, and molecular pathology. Our previous studies have described deregulation of histone deacetylases expression in ESS/UES samples. Some of these enzymes can be inhibited by substances which are already approved for treatment of cutaneous T-cell lymphoma. On the basis of published data, they may also provide a therapeutic option for ESS/UES patients. Our review focuses on molecular mechanisms of ESS/UES. It describes various aspects with special emphasis on alteration of histone deacetylation and its possible relevance for novel therapies.
Collapse
Affiliation(s)
- Andelko Hrzenjak
- Department of Internal Medicine, Division of Pulmonology, Medical University of Graz, Graz, Austria; Ludwig Boltzmann Institute for Lung Vascular Research, Medical University of Graz, Graz, Austria.
| | - Martina Dieber-Rotheneder
- Institute of Pathology, Medical University of Graz, Graz, Austria; Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Farid Moinfar
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Edgar Petru
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Kurt Zatloukal
- Institute of Pathology, Medical University of Graz, Graz, Austria
| |
Collapse
|
156
|
Quantification of vorinostat and its main metabolites in plasma and intracellular vorinostat in PBMCs by liquid chromatography coupled to tandem mass spectrometry and its relation to histone deacetylase activity in human blood. J Chromatogr B Analyt Technol Biomed Life Sci 2014; 964:212-21. [DOI: 10.1016/j.jchromb.2014.02.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 01/28/2014] [Accepted: 02/08/2014] [Indexed: 11/17/2022]
|
157
|
Zhang T, Chen Y, Li J, Yang F, Wu H, Dai F, Hu M, Lu X, Peng Y, Liu M, Zhao Y, Yi Z. Antitumor action of a novel histone deacetylase inhibitor, YF479, in breast cancer. Neoplasia 2014; 16:665-77. [PMID: 25220594 PMCID: PMC4234873 DOI: 10.1016/j.neo.2014.07.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/23/2014] [Accepted: 07/29/2014] [Indexed: 12/22/2022]
Abstract
Accumulating evidence demonstrates important roles for histone deacetylase in tumorigenesis (HDACs), highlighting them as attractive targets for antitumor drug development. Histone deactylase inhibitors (HDACIs), which have shown favorable anti-tumor activity with low toxicity in clinical investigations, are a promising class of anticancer therapeutics. Here, we screened our compound library to explore small molecules that possess anti-HDAC activity and identified a novel HDACI, YF479. Suberoylanilide hydroxamic acid (SAHA), which was the first approved HDAC inhibitor for clinical treatment by the FDA, was as positive control in our experiments. We further demonstrated YF479 abated cell viability, suppressed colony formation and tumor cell motility in vitro. To investigate YF479 with superior pharmacodynamic properties, we developed spontaneous and experimental breast cancer animal models. Our results showed YF479 significantly inhibited breast tumor growth and metastasis in vivo. Further study indicated YF479 suppressed both early and end stages of metastatic progression. Subsequent adjuvant chemotherapy animal experiment revealed the elimination of local-regional recurrence (LRR) and distant metastasis by YF479. More important, YF479 remarkably prolonged the survival of tumor-bearing mice. Intriguingly, YF479 displayed more potent anti-tumor activity in vitro and in vivo compared with SAHA. Together, our results suggest that YF479, a novel HDACI, inhibits breast tumor growth, metastasis and recurrence. In light of these results, YF479 may be an effective therapeutic option in clinical trials for patients burdened by breast cancer.
Collapse
Key Words
- hdac, histone deacetylase
- hdacis, histone deacetylase inhibitors
- saha, suberoylanilide hydroxamic acid
- lrr, local-regional recurrence
- hats, histone acetyltransferases
- vpa, valproic acid
- dapi, 4, 6-diamidino-2-phenylindole
- pcna, proliferation cell nuclear antigen
- parp, poly adp ribose polymerase
- mmp, matrix metalloproteinase
- timp, tissue inhibitor of mmp
Collapse
Affiliation(s)
- Tao Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Jingjie Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Feifei Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Haigang Wu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Fujun Dai
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Meichun Hu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Xiaoling Lu
- Biological Targeting Diagnosis and Therapy Research Center, Guangxi Medical University, 22 Shuang Yong Rd. Nanning, Guangxi 530021, China
| | - Yi Peng
- Biological Targeting Diagnosis and Therapy Research Center, Guangxi Medical University, 22 Shuang Yong Rd. Nanning, Guangxi 530021, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030.
| | - Yongxiang Zhao
- Biological Targeting Diagnosis and Therapy Research Center, Guangxi Medical University, 22 Shuang Yong Rd. Nanning, Guangxi 530021, China.
| | - Zhengfang Yi
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China.
| |
Collapse
|
158
|
Li Z, Zhu WG. Targeting histone deacetylases for cancer therapy: from molecular mechanisms to clinical implications. Int J Biol Sci 2014; 10:757-70. [PMID: 25013383 PMCID: PMC4081609 DOI: 10.7150/ijbs.9067] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 04/02/2014] [Indexed: 12/19/2022] Open
Abstract
Genetic abnormalities have been conventionally considered as hallmarks of cancer. However, studies over the past decades have demonstrated that epigenetic regulation also participates in the development of cancer. The fundamental patterns of epigenetic components, such as DNA methylation and histone modifications, are frequently altered in tumor cells. Acetylation is one of the best characterized modifications of histones, which is controlled by histone acetyltransferases (HATs) and histone deacetylases (HDACs). HDACs are a group of enzymes which catalyze the removal of the acetyl groups of both histones and non-histone proteins. HDACs are involved in modulating most key cellular processes, including transcriptional regulation, apoptosis, DNA damage repair, cell cycle control, autophagy, metabolism, senescence and chaperone function. Because HDACs have been found to function incorrectly in cancer, various HDAC inhibitors are being investigated to act as cancer chemotherapeutics. The primary purpose of this paper is to summarize recent studies of the links between HDACs and cancer, and further discuss the underlying mechanisms of anti-tumor activities of HDAC inhibitors and clinical implications.
Collapse
Affiliation(s)
- Zhiming Li
- 1. Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing 100191, China. ; 2. Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Wei-Guo Zhu
- 1. Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing 100191, China. ; 2. Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China. ; 3. Peking-Tsinghua University Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
159
|
Gao C, Bourke E, Scobie M, Famme MA, Koolmeister T, Helleday T, Eriksson LA, Lowndes NF, Brown JAL. Rational design and validation of a Tip60 histone acetyltransferase inhibitor. Sci Rep 2014; 4:5372. [PMID: 24947938 PMCID: PMC4064327 DOI: 10.1038/srep05372] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 06/02/2014] [Indexed: 12/31/2022] Open
Abstract
Histone acetylation is required for many aspects of gene regulation, genome maintenance and metabolism and dysfunctional acetylation is implicated in numerous diseases, including cancer. Acetylation is regulated by histone acetyltransferases (HATs) and histone deacetylases and currently, few general HAT inhibitors have been described. We identified the HAT Tip60 as an excellent candidate for targeted drug development, as Tip60 is a key mediator of the DNA damage response and transcriptional co-activator. Our modeling of Tip60 indicated that the active binding pocket possesses opposite charges at each end, with the positive charges attributed to two specific side chains. We used structure based drug design to develop a novel Tip60 inhibitor, TH1834, to fit this specific pocket. We demonstrate that TH1834 significantly inhibits Tip60 activity in vitro and treating cells with TH1834 results in apoptosis and increased unrepaired DNA damage (following ionizing radiation treatment) in breast cancer but not control cell lines. Furthermore, TH1834 did not affect the activity of related HAT MOF, as indicated by H4K16Ac, demonstrating specificity. The modeling and validation of the small molecule inhibitor TH1834 represents a first step towards developing additional specific, targeted inhibitors of Tip60 that may lead to further improvements in the treatment of breast cancer.
Collapse
Affiliation(s)
- Chunxia Gao
- 1] School of Chemistry, National University of Ireland Galway, Galway, Ireland [2] Department of Chemistry and Molecular Biology, University of Gothenburg, 405 30, Göteborg, Sweden [3]
| | - Emer Bourke
- 1] Discipline of Pathology, School of Medicine, Clinical Science Institute, Costello Road, National University of Ireland Galway, Galway, Ireland [2]
| | - Martin Scobie
- 1] Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden [2]
| | - Melina Arcos Famme
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Tobias Koolmeister
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Thomas Helleday
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Leif A Eriksson
- 1] School of Chemistry, National University of Ireland Galway, Galway, Ireland [2] Department of Chemistry and Molecular Biology, University of Gothenburg, 405 30, Göteborg, Sweden [3]
| | - Noel F Lowndes
- Genome Stability Laboratory, Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - James A L Brown
- 1] Genome Stability Laboratory, Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland [2] [3]
| |
Collapse
|
160
|
Pharmacophore modelling, validation, 3D virtual screening, docking, design and in silico ADMET simulation study of histone deacetylase class-1 inhibitors. Med Chem Res 2014. [DOI: 10.1007/s00044-014-1057-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
161
|
Abaza MSI, Bahman AM, Al-Attiyah RJ. Valproic acid, an anti-epileptic drug and a histone deacetylase inhibitor, in combination with proteasome inhibitors exerts antiproliferative, pro-apoptotic and chemosensitizing effects in human colorectal cancer cells: underlying molecular mechanisms. Int J Mol Med 2014; 34:513-32. [PMID: 24899129 DOI: 10.3892/ijmm.2014.1795] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 05/15/2014] [Indexed: 11/06/2022] Open
Abstract
Although the therapeutic efficacy of valproic acid (VPA) has been observed in patients with solid tumors, the very high concentration required to induce antitumor activity limits its clinical utility. The present study focused on the development of combined molecular targeted therapies using VPA and proteasome inhibitors (PIs: MG132, PI-1 and PR-39) to determine whether this combination of treatments has synergistic anticancer and chemosensitizing effects against colorectal cancer. Furthermore, the potential molecular mechanisms of action of the VPA/PI combinations were evaluated. The effects of VPA in combination with PIs on the growth of colorectal cancer cells were assessed with regard to proliferation, cell cycle, apoptosis, reactive oxygen species (ROS) generation and the expression of genes that control the cell cycle, apoptosis and pro-survival/stress-related pathways. Treatment with combinations of VPA and PIs resulted in an additive/synergistic decrease in colorectal cancer cell proliferation compared to treatment with VPA or PIs alone. The combination treatment was associated with a synergistic increase in apoptosis and in the number of cells arrested in the S phase of the cell cycle. These events were associated with increased ROS generation, pro-apoptotic gene expression and stress-related gene expression. These events were also associated with the decreased expression of anti-apoptotic genes and pro-survival genes. The combination of VPA with MG132 or PI-1 enhanced the chemosensitivity of the SW1116 (29-185‑fold) and SW837 (50-620-fold) colorectal cancer cells. By contrast, the combination of VPA/PR-39 induced a pronounced increase in the chemosensitivity of the SW837 (16-54-fold) colorectal cancer cells. These data provide a rational basis for the clinical use of this combination therapy for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Mohamed-Salah I Abaza
- Department of Biological Sciences, Faculty of Science, Kuwait University, Safat 13060, Kuwait
| | - Abdul-Majeed Bahman
- Department of Biological Sciences, Faculty of Science, Kuwait University, Safat 13060, Kuwait
| | - Raja'a J Al-Attiyah
- Department of Microbiology, Faculty of Medicine, Kuwait University, Safat 13060, Kuwait
| |
Collapse
|
162
|
Shin BS, Yoo SD, Kim TH, Bulitta JB, Landersdorfer CB, Shin JC, Choi JH, Weon KY, Joo SH, Shin S. Quantitative determination of absorption and first-pass metabolism of apicidin, a potent histone deacetylase inhibitor. Drug Metab Dispos 2014; 42:974-82. [PMID: 24644298 DOI: 10.1124/dmd.113.056713] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Apicidin, a potential oral chemotherapeutic agent, possesses potent anti-histone-deacetylase activity. After oral administration, the total bioavailability of apicidin is known to be low (14.2%-19.3%). In the present study, we evaluated the factors contributing to the low bioavailability of apicidin by means of quantitative determination of absorption fraction and first-pass metabolism after oral administration. Apicidin was given to rats by five different routes: into the femoral vein, duodenum, superior mesenteric artery, portal vein, and carotid artery. Especially, the fraction absorbed (FX) and the fraction that is not metabolized in the gut wall (FG) were separated by injection of apicidin via superior mesenteric artery, which enables bypassing the permeability barrier. The FX was 45.9% ± 9.7%, the FG was 70.9% ± 8.1% and the hepatic bioavailability (FH) was 70.6% ± 12.3%, while the pulmonary first-pass metabolism was minimal (FL = 102.8% ± 7.4%), indicating that intestinal absorption was the rate-determining step for oral absorption of apicidin. The low FX was further examined in terms of passive diffusion and transporter-mediated efflux by in vitro immobilized artificial membrane (IAM) chromatographic assay and in situ single-pass perfusion method, respectively. Although the passive diffusion potential of apicidin was high (98.01%) by the IAM assay, the in situ permeability was significantly enhanced by the presence of the P-glycoprotein (P-gp) inhibitor elacrider. These data suggest that the low bioavailability of apicidin was mainly attributed to the P-gp efflux consistent with the limited FX measured in vivo experiment.
Collapse
Affiliation(s)
- Beom Soo Shin
- College of Pharmacy, Catholic University of Daegu, Gyeongsan-si, Gyeongbuk, South Korea (B.S.S., J.C.S., J.H.C., K.Y.W., S.H.J.); School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, South Korea (S.D.Y., T.H.K.); Centre for Medicine Use and Safety, Monash University, Parkville, Australia (J.B.B., C.B.L.); Department of Pharmacy, Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk, South Korea (S.S.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Hannan JL, Kutlu O, Stopak BL, Liu X, Castiglione F, Hedlund P, Burnett AL, Bivalacqua TJ. Valproic acid prevents penile fibrosis and erectile dysfunction in cavernous nerve-injured rats. J Sex Med 2014; 11:1442-51. [PMID: 24636283 PMCID: PMC4048646 DOI: 10.1111/jsm.12522] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Bilateral cavernous nerve injury (BCNI) causes profound penile changes such as apoptosis and fibrosis leading to erectile dysfunction (ED). Histone deacetylase (HDAC) has been implicated in chronic fibrotic diseases. AIMS This study will characterize the molecular changes in penile HDAC after BCNI and determine if HDAC inhibition can prevent BCNI-induced ED and penile fibrosis. METHODS Five groups of rats (8-10 weeks, n = 10/group) were utilized: (i) sham; (ii and iii) BCNI 14 and 30 days following injury; and (iv and v) BCNI treated with HDAC inhibitor valproic acid (VPA 250 mg/kg; 14 and 30 days). All groups underwent cavernous nerve stimulation (CNS) to determine intracavernosal pressure (ICP). Penile HDAC3, HDAC4, fibronectin, and transforming growth factor-β1 (TGF-β1) protein expression (Western blot) were assessed. Trichrome staining and the fractional area of fibrosis were determined in penes from each group. Cavernous smooth muscle content was assessed by immunofluorescence to alpha smooth muscle actin (α-SMA) antibodies. MAIN OUTCOME MEASURES We measured ICP; HDAC3, HDAC4, fibronectin, and TGF-β1 protein expression; penile fibrosis; penile α-SMA content. RESULTS There was a voltage-dependent decline (P < 0.05) in ICP to CNS 14 and 30 days after BCNI. Penile HDAC3, HDAC4, and fibronectin were significantly increased (P < 0.05) 14 days after BCNI. There was a slight increase in TGF-β1 protein expression after BCNI. Histological analysis showed increased (P < 0.05) corporal fibrosis after BCNI at both time points. VPA treatment decreased (P < 0.05) penile HDAC3, HDAC4, and fibronectin protein expression as well as corporal fibrosis. There was no change in penile α-SMA between all groups. Furthermore, VPA-treated BCNI rats had improved erectile responses to CNS (P < 0.05). CONCLUSION HDAC-induced pathological signaling in response to BCNI contributes to penile vascular dysfunction. Pharmacological inhibition of HDAC prevents penile fibrosis, normalizes fibronectin expression, and preserves erectile function. The HDAC pathway may represent a suitable target in preventing the progression of ED occurring post-radical prostatectomy.
Collapse
Affiliation(s)
- Johanna L. Hannan
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Omer Kutlu
- Department of Urology, Karadeniz Technical University, Trabzon, Turkey
| | - Bernard L. Stopak
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Xiaopu Liu
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Fabio Castiglione
- Department of Urology, Urological Research Institute, San Raffaele Research Institute, Milan, Italy
| | - Petter Hedlund
- Department of Urology, Urological Research Institute, San Raffaele Research Institute, Milan, Italy
- Department of Clinical Pharmacology, Linköping University, Linköping, Sweden
| | - Arthur L. Burnett
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Trinity J. Bivalacqua
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
164
|
CAI LISHENG, MA XUDONG, HUANG YIQUN, ZOU YONG, CHEN XINGSHENG. Aberrant histone methylation and the effect of Suv39H1 siRNA on gastric carcinoma. Oncol Rep 2014; 31:2593-600. [DOI: 10.3892/or.2014.3135] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 03/21/2014] [Indexed: 11/05/2022] Open
|
165
|
A histone deacetylase inhibitor YCW1 with antitumor and antimetastasis properties enhances cisplatin activity against non-small cell lung cancer in preclinical studies. Cancer Lett 2014; 346:84-93. [DOI: 10.1016/j.canlet.2013.12.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 12/04/2013] [Accepted: 12/09/2013] [Indexed: 01/20/2023]
|
166
|
Bloch KM, Sills GJ, Pirmohamed M, Alfirevic A. Pharmacogenetics of antiepileptic drug-induced hypersensitivity. Pharmacogenomics 2014; 15:857-68. [PMID: 24897291 DOI: 10.2217/pgs.14.65] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Antiepileptic drugs can induce potentially life-threatening hypersensitivity reactions such as Stevens–Johnson syndrome at a frequency of one in 10,000 to one in 1000 treated patients. There is a considerable cross-reactivity among different antiepileptic drugs but the mechanisms are not known. In this review we have summarized current evidence on antiepileptic drug-induced hypersensitivity reactions and performed meta-analyses of published case–control studies that investigated associations between HLA alleles and several antiepileptic drugs in diverse populations. As the heterogeneity between studies was high, we conducted subsequent subgroup analyses and showed that HLA-B*15:02 was associated with carbamazepine, lamotrigine and phenytoin-induced Stevens–Johnson syndrome in Asian populations indicating that pretreatment testing may prevent cross-reactivity. Additionally, we explored the potential of new, high-throughput technologies that may help to understand the mechanisms and predict the risk of adverse drug reactions in the future.
Collapse
Affiliation(s)
- Katarzyna M Bloch
- The Wolfson Centre for Personalised Medicine, Department of Molecular & Clinical Pharmacology, University of Liverpool, Block A: Waterhouse Buildings, 1–5 Brownlow Street, Liverpool, L69 3GL, UK
| | - Graeme J Sills
- The Wolfson Centre for Personalised Medicine, Department of Molecular & Clinical Pharmacology, University of Liverpool, Block A: Waterhouse Buildings, 1–5 Brownlow Street, Liverpool, L69 3GL, UK
| | - Munir Pirmohamed
- The Wolfson Centre for Personalised Medicine, Department of Molecular & Clinical Pharmacology, University of Liverpool, Block A: Waterhouse Buildings, 1–5 Brownlow Street, Liverpool, L69 3GL, UK
| | - Ana Alfirevic
- The Wolfson Centre for Personalised Medicine, Department of Molecular & Clinical Pharmacology, University of Liverpool, Block A: Waterhouse Buildings, 1–5 Brownlow Street, Liverpool, L69 3GL, UK
| |
Collapse
|
167
|
Regna NL, Chafin CB, Hammond SE, Puthiyaveetil AG, Caudell DL, Reilly CM. Class I and II histone deacetylase inhibition by ITF2357 reduces SLE pathogenesis in vivo. Clin Immunol 2014; 151:29-42. [PMID: 24503172 PMCID: PMC3963170 DOI: 10.1016/j.clim.2014.01.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 01/02/2014] [Accepted: 01/04/2014] [Indexed: 12/24/2022]
Abstract
We sought to determine if a specific class I and II HDAC inhibitor (ITF2357) was able to decrease disease in lupus-prone NZB/W mice through regulation of T cell profiles. From 22 to 38 weeks-of-age, NZB/W and non-lupus NZW mice were treated with ITF2357 (5 mg/kg or 10 mg/kg), or vehicle control. Body weight and proteinuria were measured every 2 weeks, while sera anti-dsDNA and cytokine levels were measured every 4 weeks. Kidney disease was determined by sera IgG levels, immune complex deposition, and renal pathology. T lymphocyte profiles were assessed using flow cytometric analyses. Our results showed that NZB/W mice treated with the 10 mg/kgof ITF2357 had decreased renal disease and inflammatory cytokines in the sera. Treatment with ITF2357 decreased the Th17 phenotype while increasing the percentage of Tregs as well as Foxp3 acetylation. These results suggest that specific HDAC inhibition may decrease disease by altering T cell differentiation and acetylation.
Collapse
Affiliation(s)
- Nicole L Regna
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg VA, 24061, USA.
| | - Cristen B Chafin
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg VA, 24061, USA
| | - Sarah E Hammond
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg VA, 24061, USA
| | - Abdul G Puthiyaveetil
- Department of Biotechnology, American University of Ras Al Khaimah, PO Box 10021, United Arab Emirates
| | - David L Caudell
- Department of Pathology, Center for Comparative Medicine Research, Wake Forest School of Medicine, Winston-Salem NC 27157, USA
| | - Christopher M Reilly
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg VA, 24061, USA; Edward Via College of Osteopathic Medicine, Blacksburg, VA 24060, USA
| |
Collapse
|
168
|
Okabe S, Tauchi T, Kimura S, Maekawa T, Kitahara T, Tanaka Y, Ohyashiki K. Combining the ABL1 kinase inhibitor ponatinib and the histone deacetylase inhibitor vorinostat: a potential treatment for BCR-ABL-positive leukemia. PLoS One 2014; 9:e89080. [PMID: 24586514 PMCID: PMC3938434 DOI: 10.1371/journal.pone.0089080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 01/15/2014] [Indexed: 11/19/2022] Open
Abstract
Resistance to imatinib (Gleevec®) in cancer cells is frequently because of acquired point mutations in the kinase domain of BCR-ABL. Ponatinib, also known as AP24534, is an oral multi-targeted tyrosine kinase inhibitor (TKI), and it has been investigated in a pivotal phase 2 clinical trial. The histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid) has been evaluated for its significant clinical activity in hematological malignancies. Thus, treatments combining ABL TKIs with additional drugs may be a promising strategy in the treatment of leukemia. In the current study, we analyzed the efficacy of ponatinib and vorinostat treatment by using BCR-ABL-positive cell lines. Treatment with ponatinib for 72 h inhibited cell growth and induced apoptosis in K562 cells in a dose-dependent manner. We found that ponatinib potently inhibited the growth of Ba/F3 cells ectopically expressing BCR-ABL T315I mutation. Upon BCR-ABL phosphorylation, Crk-L was decreased, and poly (ADP-ribose) polymerase (PARP) was activated in a dose-dependent manner. Combined treatment of Ba/F3 T315I mutant cells with vorinostat and ponatinib resulted in significantly increased cytotoxicity. Additionally, the intracellular signaling of ponatinib and vorinostat was examined. Caspase 3 and PARP activation increased after combination treatment with ponatinib and vorinostat. Moreover, an increase in the phosphorylation levels of γH2A.X was observed. Previously established ponatinib-resistant Ba/F3 cells were also resistant to imatinib, nilotinib, and dasatinib. We investigated the difference in the efficacy of ponatinib and vorinostat by using ponatinib-resistant Ba/F3 cells. Combined treatment of ponatinib-resistant cells with ponatinib and vorinostat caused a significant increase in cytotoxicity. Thus, combined administration of ponatinib and vorinostat may be a powerful strategy against BCR-ABL mutant cells and could enhance the cytotoxic effects of ponatinib in those BCR-ABL mutant cells.
Collapse
Affiliation(s)
- Seiichi Okabe
- First Department of Internal Medicine, Tokyo Medical University, Tokyo, Japan
- * E-mail:
| | - Tetsuzo Tauchi
- First Department of Internal Medicine, Tokyo Medical University, Tokyo, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Taira Maekawa
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
| | - Toshihiko Kitahara
- First Department of Internal Medicine, Tokyo Medical University, Tokyo, Japan
| | - Yoko Tanaka
- First Department of Internal Medicine, Tokyo Medical University, Tokyo, Japan
| | - Kazuma Ohyashiki
- First Department of Internal Medicine, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
169
|
Ferreira ACDS, Robaina MC, Rezende LMMD, Severino P, Klumb CE. Histone deacetylase inhibitor prevents cell growth in Burkitt's lymphoma by regulating PI3K/Akt pathways and leads to upregulation of miR-143, miR-145, and miR-101. Ann Hematol 2014; 93:983-93. [PMID: 24577510 DOI: 10.1007/s00277-014-2021-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 01/22/2014] [Indexed: 01/31/2023]
Abstract
Burkitt lymphoma (BL) is an aggressive B-cell lymphoma more common in children comprising one third of pediatric non-Hodgkin lymphoma cases. The recent discovery in BL pathogenesis highlighted the activation of PI3K pathway in cooperation with Myc in the development of BL. In this study, we demonstrated that PI3K/Akt pathway is a target to histone deacetylase inhibitor (HDACi) in BL cells. The combination of HDACi (sodium butyrate, NaB) and chemotherapy (VP-16) inhibited 51 % of the proliferation and enhanced the blockage of the cell cycle progression at G2/M with a concurrent decrease in the S phase. Microarray profiling showed a synergistic action of NaB/VP-16 combination through the differential regulation of 1,413 genes. Comparing VP-16 treatment with the NaB/VP-16 combination, 318 genes were deregulated: 250 genes were downregulated, and 68 were upregulated when compared with untreated cells. Among these genes, six (CDKN1A, CCND1, FAS, CHEK2, MDM4, and SESN2) belong to the p53-signaling pathway. The activation of this signaling pathway is usually induced by stress signals and ultimately leads to cell cycle arrest. Besides, the inhibition of the cell growth was related to reduced Akt phosphorylation, and decrease of c-Myc protein expression by about 60 % (p ≤ 0.005). Moreover, HDACi enhanced miR-101, miR-143, and miR-145 levels in BL cell line, which were inversely associated with the levels of miR-101, miR-143, and miR-145 found to be extremely downregulated in the sample of BL patients. We highlight the fact that effective combinations of HDACis with other target drugs could improve BL therapy in the future.
Collapse
Affiliation(s)
- Ana Carolina dos Santos Ferreira
- Programa de Pesquisa em Hemato-Oncologia Molecular, Coordenação Geral Técnico-Científica, Instituto Nacional de Câncer-INCA, Rio de Janeiro, Brazil
| | | | | | | | | |
Collapse
|
170
|
Maleszewska M, Steranka A, Kaminska B. The effects of selected inhibitors of histone modifying enzyme on C6 glioma cells. Pharmacol Rep 2014; 66:107-13. [PMID: 24905315 DOI: 10.1016/j.pharep.2013.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 05/13/2013] [Accepted: 08/20/2013] [Indexed: 10/25/2022]
Abstract
BACKGROUND Aberrant epigenetic histone modifications are implicated in cancer pathobiology, therefore histone modifying enzymes are emerging targets for anti-cancer therapy. There is a few evidence for deregulation of the histone modifying enzymes in glioblastomas. Glioma treatment is a clinical challenge due to its resistance to current therapies. METHODS The effect of selected inhibitors on epigenetic modifications and viability of glioma C6 cells were studied using immunofluorescence and MTT metabolism test. RESULTS We found that VPA and TSA increase histone H4 acetylation in glioma cells, while chaetocin and BIX01294 at low concentrations reduce H3K9me3, and 3DZNep decreases H3K27me3. Long-term treatment with some epigenetic inhibitors affects viability of glioma cells. CONCLUSIONS We established the concentrations of selected inhibitors which in C6 glioma cells inhibit the enzyme activity, but do not decrease cell viability, hence allow to study the role of histone modifications in C6 glioma biology.
Collapse
Affiliation(s)
- Marta Maleszewska
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology, Warszawa, Poland
| | - Aleksandra Steranka
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology, Warszawa, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology, Warszawa, Poland.
| |
Collapse
|
171
|
Dalton VS, Kolshus E, McLoughlin DM. Epigenetics and depression: return of the repressed. J Affect Disord 2014; 155:1-12. [PMID: 24238955 DOI: 10.1016/j.jad.2013.10.028] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/15/2013] [Accepted: 10/16/2013] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Epigenetics has recently emerged as a potential mechanism by which adverse environmental stimuli can result in persistent changes in gene expression. Epigenetic mechanisms function alongside the DNA sequence to modulate gene expression and ultimately influence protein production. The current review provides an introduction and overview of epigenetics with a particular focus on preclinical and clinical studies relevant to major depressive disorder (MDD). METHODS PubMed and Web of Science databases were interrogated from January 1995 up to December 2012 using combinations of search terms, including "epigenetic", "microRNA" and "DNA methylation" cross referenced with "depression", "early life stress" and "antidepressant". RESULTS There is an association between adverse environmental stimuli, such as early life stress, and epigenetic modification of gene expression. Epigenetic changes have been reported in humans with MDD and may serve as biomarkers to improve diagnosis. Antidepressant treatments appear to reverse or initiate compensatory epigenetic alterations that may be relevant to their mechanism of action. LIMITATIONS As a narrative review, the current report was interpretive and qualitative in nature. CONCLUSION Epigenetic modification of gene expression provides a mechanism for understanding the link between long-term effects of adverse life events and the changes in gene expression that are associated with depression. Although still a developing field, in the future, epigenetic modifications of gene expression may provide novel biomarkers to predict future susceptibility and/or onset of MDD, improve diagnosis, and aid in the development of epigenetics-based therapies for depression.
Collapse
Affiliation(s)
- Victoria S Dalton
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland; Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James's Street, Dublin 8, Ireland
| | - Erik Kolshus
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland; Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James's Street, Dublin 8, Ireland
| | - Declan M McLoughlin
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland; Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James's Street, Dublin 8, Ireland.
| |
Collapse
|
172
|
Ganai SA, Shanmugam K, Mahadevan V. Energy-optimised pharmacophore approach to identify potential hotspots during inhibition of Class II HDAC isoforms. J Biomol Struct Dyn 2014; 33:374-87. [PMID: 24460542 DOI: 10.1080/07391102.2013.879073] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Histone deacetylases (HDACs) are conjugated enzymes that modulate chromatin architecture by deacetylating lysine residues on the histone tails leading to transcriptional repression. Pharmacological interventions of these enzymes with small molecule inhibitors called Histone deacetylase inhibitors (HDACi) have shown enhanced acetylation of the genome and are hence emerging as potential targets at the clinic. Type-specific inhibition of Class II HDACs has shown enhanced therapeutic benefits against developmental and neurodegenerative disorders. However, the structural identity of class-specific isoforms limits the potential of their inhibitors in precise targeting of their enzymes. Diverse strategies have been implemented to recognise the features in HDAC enzymes which may help in identifying isoform specificity factors. This work attempts a computational approach that combines in silico docking and energy-optimised pharmacophore (E-pharmacophore) mapping of 18 known HDAC inhibitors and has identified structural variations that regulate their interactions against the six Class II HDAC enzymes considered for the study. This combined approach establishes that inhibitors possessing higher number of aromatic rings in different structural regions might function as potent inhibitors, while inhibitors with scarce ring structures might point to compromised potency. This would aid the rationale for chemical optimisation and design of isoform selective HDAC inhibitors with enhanced affinity and therapeutic efficiency.
Collapse
Affiliation(s)
- Shabir Ahmad Ganai
- a Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical & Biotechnology , SASTRA University , Thanjavur 613401 , India
| | | | | |
Collapse
|
173
|
Coiffier B, Pro B, Prince HM, Foss F, Sokol L, Greenwood M, Caballero D, Morschhauser F, Wilhelm M, Pinter-Brown L, Padmanabhan Iyer S, Shustov A, Nielsen T, Nichols J, Wolfson J, Balser B, Horwitz S. Romidepsin for the treatment of relapsed/refractory peripheral T-cell lymphoma: pivotal study update demonstrates durable responses. J Hematol Oncol 2014; 7:11. [PMID: 24456586 PMCID: PMC4016573 DOI: 10.1186/1756-8722-7-11] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 01/18/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Romidepsin is a structurally unique, potent, bicyclic class 1 selective histone deacetylase inhibitor approved by the US Food and Drug Administration for the treatment of patients with cutaneous T-cell lymphoma who have received ≥ 1 prior systemic therapy and patients with peripheral T-cell lymphoma (PTCL) who have received ≥ 1 prior therapy. Approval for PTCL was based on results (n = 130; median follow-up, 13.4 months) from the pivotal study of romidepsin for the treatment of relapsed/refractory PTCL. The objective is to present updated data (median follow-up, 22.3 months) and to characterize patients who achieved long-term responses (≥ 12 months) to romidepsin. METHODS Patients with PTCL who relapsed from or were refractory to ≥ 1 prior systemic therapy received romidepsin 14 mg/m2 as a 4-hour intravenous infusion on days 1, 8, and 15 every 28 days for up to 6 cycles; patients with response or stable disease could continue romidepsin beyond 6 cycles. The primary endpoint was rate of confirmed/unconfirmed complete response (CR/CRu) determined by an Independent Review Committee. Secondary endpoints included objective response rate (ORR) and duration of response (DOR). For patients who achieved CR/CRu, baseline characteristics by DOR (≥ 12 vs < 12 months) were examined. RESULTS The ORR to romidepsin was 25%, including 15% with CR/CRu. The median DOR for all responders was 28 months (range, < 1-48+) and was not reached for those who achieved CR/CRu. Patients with lack of response or transient response to prior therapy achieved durable responses with romidepsin. Of the 19 patients who achieved CR/CRu, 10 had long-term (≥ 12 months) responses; none of the baseline characteristics examined-including heavy pretreatment, response to prior therapy, or advanced disease-precluded long-term responses to romidepsin. With a median progression-free survival of 29 months, patients who achieved CR/CRu for ≥ 12 months had significantly longer survival vs those with CR/CRu for < 12 months or < CR/CRu. Extended treatment and longer follow-up did not affect the reported safety profile of romidepsin. CONCLUSIONS Treatment with romidepsin leads to highly durable responses in a subset of patients with relapsed/refractory PTCL, with responses ongoing as long as 48 months.
Collapse
|
174
|
Autism’s cancer connection: The anti-proliferation hypothesis and why it may matter. Med Hypotheses 2014; 82:26-35. [DOI: 10.1016/j.mehy.2013.10.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/27/2013] [Accepted: 10/31/2013] [Indexed: 12/30/2022]
|
175
|
Biemar F, Foti M. Global progress against cancer-challenges and opportunities. Cancer Biol Med 2013; 10:183-6. [PMID: 24349827 PMCID: PMC3860343 DOI: 10.7497/j.issn.2095-3941.2013.04.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 10/16/2013] [Indexed: 12/13/2022] Open
Abstract
The last ten years have seen remarkable progress in cancer research. However, despite significant breakthroughs in the understanding, prevention, and treatment of cancer, the disease continues to affect millions of people worldwide. Cancer’s complexity compounded with financial, policy and regulatory roadblocks has slowed the rate of progress being made against cancer. In this paper, we review a few of the most recent breakthroughs that are fueling medical advances and bringing new hope for patients affected by this devastating disease. We also address the challenges facing us and the opportunities to accelerate future progress against cancer. The efforts of the American Association for Cancer Research (AACR) to address the cancer burden already extend beyond the borders of the United States of America. The AACR is committed to increasing its efforts to stem the tide of cancer worldwide by promoting innovative programs, strategies, and initiatives for cancer researchers and all those engaged in cancer-related biomedical sciences around the world.
Collapse
Affiliation(s)
- Frédéric Biemar
- American Association for Cancer Research, Philadelphia, PA 19130, USA
| | - Margaret Foti
- American Association for Cancer Research, Philadelphia, PA 19130, USA
| |
Collapse
|
176
|
Zhang Z, Convertini P, Shen M, Xu X, Lemoine F, de la Grange P, Andres DA, Stamm S. Valproic acid causes proteasomal degradation of DICER and influences miRNA expression. PLoS One 2013; 8:e82895. [PMID: 24358235 PMCID: PMC3866160 DOI: 10.1371/journal.pone.0082895] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 11/07/2013] [Indexed: 11/18/2022] Open
Abstract
Valproic acid (VPA) is a commonly used drug to treat epilepsy and bipolar disorders. Known properties of VPA are inhibitions of histone deacetylases and activation of extracellular signal regulated kinases (ERK), which cannot fully explain VPA's clinical features. We found that VPA induces the proteasomal degradation of DICER, a key protein in the generation of micro RNAs. Unexpectedly, the concentration of several micro RNAs increases after VPA treatment, which is caused by the upregulation of their hosting genes prior to DICER degradation. The data suggest that a loss of DICER protein and changes in micro RNA concentration contributes to the clinical properties of VPA. VPA can be used experimentally to down regulate DICER protein levels, which likely reflects a natural regulation of DICER.
Collapse
Affiliation(s)
- Zhaiyi Zhang
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Paolo Convertini
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Manli Shen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Xiu Xu
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Frédéric Lemoine
- GenoSplice Technology, Hôpital Saint-Louis, Av Claude Vellefaux, Paris, France
| | - Pierre de la Grange
- GenoSplice Technology, Hôpital Saint-Louis, Av Claude Vellefaux, Paris, France
| | - Douglas A. Andres
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Stefan Stamm
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
177
|
Abstract
PURPOSE The pharmacology, pharmacokinetic and pharmacodynamic properties, and clinical data on a novel therapy for the treatment of cutaneous or peripheral T-cell lymphoma (CTCL, PTCL) are summarized. SUMMARY Romidepsin is the only bicyclic histone deacetylase (HDAC) inhibitor to undergo clinical development. A potent and specific inhibitor of class 1 HDACs, romidepsin has linear pharmacokinetics and is primarily metabolized by cytochrome P-450 isoenzyme 3A4. In two Phase II studies involving patients with relapsed or refractory CTCL, romidepsin therapy produced overall response rates of 34-35% (including patients with advanced and heavily pretreated disease), with a complete response seen in about 6% of patients in both studies; romidepsin responses were seen across all evaluated disease sites (skin, blood, lymph, viscera). In two Phase II studies in patients with relapsed or refractory PTCL, romidepsin produced overall response rates of 25-38%, and 15-18% of patients experienced a complete response; therapeutic responses were seen across major PTCL subtypes regardless of the number or types of previous therapies or refractoriness to the last prior therapy. In clinical trials to date, romidepsin therapy was generally well tolerated, with nausea, fatigue, and vomiting reported as the most common nonhematologic adverse events. However, thrombocytopenia and neutropenia are relatively common events, especially in patients with PTCL. CONCLUSION Romidepsin, a class 1-specific HDAC inhibitor, induces durable responses, with a manageable toxicity profile, in patients with relapsed or refractory CTCL or PTCL who have few therapeutic options.
Collapse
Affiliation(s)
- Angie L McGraw
- Middle Tennessee Medical Center, 1700 Medical Center Parkway, Murfreesboro, TN 37129, USA.
| |
Collapse
|
178
|
Salahshoor MR, Dastjerdi MN, Jalili C, Mardani M, Khazaei M, Darehdor AS, Valiani A, Roshankhah S. Combination of Salermide and Cholera Toxin B Induce Apoptosis in MCF-7 but Not in MRC-5 Cell Lines. Int J Prev Med 2013; 4:1402-13. [PMID: 24498496 PMCID: PMC3898446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 05/28/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Sirtuin1 is an enzyme that deacetylates histones and several non-histone proteins including P53 during the stress. P300 is a member of the histone acetyl transferase family and enzyme that acetylates histones. Hereby, this study describes the potency combination of Salermide as a Sirtuin1 inhibitor and cholera toxin B (CTB) as a P300 activator to induce apoptosis Michigan Cancer Foundation-7 (MCF-7) and MRC-5. METHODS Cells were cultured and treated with a combination of Salermide and CTB respectively at concentrations of 80.56 and 85.43 μmol/L based on inhibitory concentration 50 indexes at different times. The percentage of apoptotic cells were measured by flow cytometry. Real-time polymerase chain reaction was performed to estimate the messenger ribonucleic acid expression of Sirtuin1 and P300 in cells. Enzyme linked immunosorbent assay and Bradford protein techniques were used to detect the endogenous levels of total and acetylated P53 protein generated in both cell lines. RESULTS Our findings indicated that the combination of two drugs could effectively induced apoptosis in MCF-7 significantly higher than MRC-5. We showed that expression of Sirtuin1 and P300 was dramatically down-regulated with increasing time by the combination of Salermide and CTB treatment in MCF-7, but not MRC-5. The acetylated and total P53 protein levels were increased more in MCF-7 than MRC-5 with incubated combination of drugs at different times. Combination of CTB and Salermide in 72 h through decreasing expression of Sirtuin1 and P300 genes induced acetylation of P53 protein and consequently showed the most apoptosis in MCF-7 cells, but it could be well-tolerated in MRC-5. CONCLUSION Therefore, combination of drugs could be used as an anticancer agent.
Collapse
Affiliation(s)
- Mohammad Reza Salahshoor
- Fertility and Infertility Research Center, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehdi Nikbakht Dastjerdi
- Department of Anatomical Sciences and Molecular Biology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran,Correspondence to: Dr. Mehdi Nikbakht Dastjerdi, Department of Anatomical Sciences and Molecular Biology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran. E-mail:
| | - Cyrus Jalili
- Department of Anatomical Sciences and Molecular Biology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Mardani
- Department of Anatomical Sciences and Molecular Biology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ahmad Shabanizadeh Darehdor
- Department of Anatomical Sciences and Molecular Biology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Valiani
- Fertility and Infertility Research Center, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shiva Roshankhah
- Fertility and Infertility Research Center, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
179
|
Baldo BA, Pham NH. Adverse reactions to targeted and non-targeted chemotherapeutic drugs with emphasis on hypersensitivity responses and the invasive metastatic switch. Cancer Metastasis Rev 2013; 32:723-61. [PMID: 24043487 PMCID: PMC7102343 DOI: 10.1007/s10555-013-9447-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
More than 100 drugs are used to treat the many different cancers. They can be divided into agents with relatively broad, non-targeted specificity and targeted drugs developed on the basis of a more refined understanding of individual cancers and directed at specific molecular targets on different cancer cells. Individual drugs in both groups have been classified on the basis of their mechanism of action in killing cancer cells. The targeted drugs include proteasome inhibitors, toxic chimeric proteins and signal transduction inhibitors such as tyrosine kinase (non-receptor and receptor), serine/threonine kinase, histone deacetylase and mammalian target of rapamycin inhibitors. Increasingly used targeted vascular (VEGF) and platelet-derived endothelial growth factor blockade can provoke a range of pathological consequences. Many of the non-targeted drugs are cytotoxic, suppressing haematopoiesis as well as provoking cutaneous eruptions and vascular, lung and liver injury. Cytotoxic side effects of the targeted drugs occur less often and usually with less severity, but they show their own unusual adverse effects including, for example, a lengthened QT interval, a characteristic papulopustular rash, nail disorders and a hand-foot skin reaction variant. The term hypersensitivity is widely used across a number of disciplines but not always with the same definition in mind, and the terminology needs to be standardised. This is particularly apparent in cancer chemotherapy where anti-neoplastic drug-induced thrombocytopenia, neutropenia, anaemia, vascular disorders, liver injury and lung disease as well as many dermatological manifestations sometimes have an immune basis. The most insidious of all adverse consequences of targeted therapies, however, are tumour adaptation, increased malignancy and the invasive metastatic switch seen with anti-angiogenic drugs that inhibit the VEGF-A pathway. Adverse reactions to 44 non-targeted and 33 targeted, frequently used, chemotherapeutic drugs are presented together with discussions of diagnosis, premedications, desensitizations and importance of understanding the mechanisms underlying the various drug-induced reactions. There is need for wide-ranging acceptance of what constitutes a hypersensitivity reaction and for allergists to be more involved in the diagnosis, treatment and prevention of chemotherapeutic drug-induced hypersensitivity reactions.
Collapse
Affiliation(s)
- Brian A Baldo
- Molecular Immunology Unit, Kolling Institute of Medical Research, Royal North Shore Hospital of Sydney, Sydney, New South Wales, Australia,
| | | |
Collapse
|
180
|
Deng L, Song Y, Zhu J, Zheng W, Wang X, Xie Y, Lin N, Tu M, Ping L, Ying Z, Liu W, Zhang C. Secondary central nervous system involvement in 599 patients with diffuse large B-cell lymphoma: are there any changes in the rituximab era? Int J Hematol 2013; 98:664-71. [DOI: 10.1007/s12185-013-1458-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 10/29/2013] [Accepted: 10/29/2013] [Indexed: 01/18/2023]
|
181
|
Seuter S, Pehkonen P, Heikkinen S, Carlberg C. Dynamics of 1α,25-dihydroxyvitamin D3-dependent chromatin accessibility of early vitamin D receptor target genes. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:1266-75. [PMID: 24185200 DOI: 10.1016/j.bbagrm.2013.10.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 10/01/2013] [Accepted: 10/18/2013] [Indexed: 01/05/2023]
Abstract
The signaling cascade of the transcription factor vitamin D receptor (VDR) is triggered by its specific ligand 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3). In this study we demonstrate that in THP-1 human monocytic leukemia cells 87.4% of the 1034 most prominent genome-wide VDR binding sites co-localize with loci of open chromatin. At 165 of them 1α,25(OH)2D3 strongly increases chromatin accessibility and has at further 217 sites weaker effects. Interestingly, VDR binding sites in 1α,25(OH)2D3-responsive chromatin regions are far more often composed of direct repeats with 3 intervening nucleotides (DR3s) than those in ligand insensitive regions. DR3-containing VDR sites are enriched in the neighborhood of genes that are involved in controling cellular growth, while non-DR3 VDR binding is often found close to genes related to immunity. At the example of six early VDR target genes we show that the slope of their 1α,25(OH)2D3-induced transcription correlates with the basal chromatin accessibility of their major VDR binding regions. However, the chromatin loci controlling these genes are indistinguishable in their VDR association kinetics. Taken together, ligand responsive chromatin loci represent dynamically regulated contact points of VDR with the genome, from where it controls early 1α,25(OH)2D3 target genes.
Collapse
Key Words
- 1α,25(OH)(2)D(3) or 1,25D
- 1α,25-dihydroxyvitamin D(3)
- B2M
- CAMP
- CD14
- ChIP
- ChIP sequencing
- ChIP-seq
- DR3
- FAIRE
- FAIRE sequencing
- FAIRE-seq
- FANCE
- FBP1
- FCS
- FDR
- Fanconi anemia, complementation group E
- GAPDH
- GEO
- GREAT
- Gene Expression Omnibus
- Genomic Regions Enrichment of Annotations Tool
- H3K27ac
- HBB
- HBEGF
- HDAC
- HDAC inhibitor
- HPRTI
- IGV
- Integrative Genomics Viewer
- MB
- NFKBIA
- NINJ1
- Open chromatin
- PDCD1LG2
- PPARGC1B
- PSMA6
- SAHA
- SCTR
- SEMA6B
- TMEM37
- TSS
- TsA
- VDR
- VPA
- Vitamin D
- beta-2-microglobulin
- cathelicidin antimicrobial peptide
- chromatin immunoprecipitation
- cluster of differentiation 14 molecule
- direct repeat spaced by 3 nucleotides
- false discovery rate
- fetal calf serum
- formaldehyde-assisted isolation of regulatory elements
- fructose-1,6-bisphosphatase 1
- gylcerinaldehyde-3-phosphate-dehydrogenase
- hemoglobin, beta
- heparin-binding EGF-like growth factor
- histone 3 acetylation at lysine 27
- histone deacetylase
- hypoxanthine phosphoribosyltransferase 1
- myoglobin
- ninjurin 1
- nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha
- peroxisome proliferator-activated receptor γ, co-activator 1β
- programmed cell death 1 ligand 2
- proteasome subunit, alpha type, 6
- qPCR
- real-time quantitative polymerase chain reaction
- secretin receptor
- semaphorin 6B
- suberoylanilide hydroxamic acid
- transcription start site
- transmembrane protein 37
- trichostatin A
- valproic acid
- vitamin D receptor
Collapse
Affiliation(s)
- Sabine Seuter
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | | | | | | |
Collapse
|
182
|
Omene C, Kalac M, Wu J, Marchi E, Frenkel K, O’Connor OA. Propolis and its Active Component, Caffeic Acid Phenethyl Ester (CAPE), Modulate Breast Cancer Therapeutic Targets via an Epigenetically Mediated Mechanism of Action. JOURNAL OF CANCER SCIENCE & THERAPY 2013; 5:334-342. [PMID: 24466386 PMCID: PMC3898618] [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
Alternative remedies for cancer treatment is a multi-billion dollar industry. In particular, breast cancer (BC) patients use alternative and natural remedies more frequently than patients with other malignancies. Propolis is an example of a honeybee-produced naturopathic formulation, contents of which differ by geographic location. It is readily available, affordable, and in use safely since ancient times globally. Caffeic acid phenethyl ester (CAPE) is a major active component in propolis and is thought to be responsible for its varied properties, including antibacterial, antiviral, antifungal, antioxidant, anti-inflammatory and anticancer. CAPE is effective in many models of human cancer, including BC as we have previously shown. CAPE affects genes associated with tumor cell growth and survival, angiogenesis and chemoresistance. We demonstrate that these are related in part to CAPE's role as a histone deacetylase inhibitor, a class of drugs designated as epigenetic agents that modulate the activities of oncogenes and tumor suppressor genes. CAPE and propolis, cause an accumulation of acetylated histone proteins in MCF-7 (ER+) and MDA-MB-231 (ER-/PR-/Her2-) cells with associated decreases in ER and PR in MCF-7 cells, and upregulation of ER and decrease in EGFR in MDA-231 cells. In addition, these products reduced activated phosphorylated Her2 protein in SKBR3 (Her2 +) cells. Interestingly, propolis, when normalized for CAPE content, appears to be more potent than CAPE alone similarly to the greater effects of complete foods than isolated components. These data provide a potential mechanistic basis for one of the oldest naturopathic agents used in medicine and cancer treatment.
Collapse
Affiliation(s)
- Coral Omene
- Department of Medicine, NYU School of Medicine, New York, USA
- NYU Cancer Institute, NYU School of Medicine, New York, USA
| | - Matko Kalac
- Department of Medicine, NYU School of Medicine, New York, USA
- NYU Cancer Institute, NYU School of Medicine, New York, USA
| | - Jing Wu
- Department of Environmental Medicine, NYU School of Medicine, New York, USA
| | - Enrica Marchi
- Department of Medicine, NYU School of Medicine, New York, USA
- Center for Lymphoid Malignancies, Columbia University Medical Center, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, USA
| | - Krystyna Frenkel
- Department of Environmental Medicine, NYU School of Medicine, New York, USA
- NYU Cancer Institute, NYU School of Medicine, New York, USA
| | - Owen A O’Connor
- Department of Medicine, NYU School of Medicine, New York, USA
- Center for Lymphoid Malignancies, Columbia University Medical Center, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, USA
| |
Collapse
|
183
|
Ryynänen J, Seuter S, Campbell MJ, Carlberg C. Gene regulatory scenarios of primary 1,25-dihydroxyvitamin d3 target genes in a human myeloid leukemia cell line. Cancers (Basel) 2013; 5:1221-41. [PMID: 24202443 PMCID: PMC3875937 DOI: 10.3390/cancers5041221] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 09/19/2013] [Accepted: 09/26/2013] [Indexed: 12/14/2022] Open
Abstract
Genome- and transcriptome-wide data has significantly increased the amount of available information about primary 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) target genes in cancer cell models, such as human THP-1 myelomonocytic leukemia cells. In this study, we investigated the genes G0S2, CDKN1A and MYC as master examples of primary vitamin D receptor (VDR) targets being involved in the control of cellular proliferation. The chromosomal domains of G0S2 and CDKN1A are 140-170 kb in size and contain one and three VDR binding sites, respectively. This is rather compact compared to the MYC locus that is 15 times larger and accommodates four VDR binding sites. All eight VDR binding sites were studied by chromatin immunoprecipitation in THP-1 cells. Interestingly, the site closest to the transcription start site of the down-regulated MYC gene showed 1,25(OH)2D3-dependent reduction of VDR binding and is not associated with open chromatin. Four of the other seven VDR binding regions contain a typical DR3-type VDR binding sequence, three of which are also occupied with VDR in macrophage-like cells. In conclusion, the three examples suggest that each VDR target gene has an individual regulatory scenario. However, some general components of these scenarios may be useful for the development of new therapy regimens.
Collapse
Affiliation(s)
- Jussi Ryynänen
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, POB 1627, Kuopio FI-70211, Finland; E-Mails: (J.R.), (S.S.)
| | - Sabine Seuter
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, POB 1627, Kuopio FI-70211, Finland; E-Mails: (J.R.), (S.S.)
| | - Moray J. Campbell
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA; E-Mail:
| | - Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, POB 1627, Kuopio FI-70211, Finland; E-Mails: (J.R.), (S.S.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +358-40-355-3062
| |
Collapse
|
184
|
Wang D, Zhang H, Li M, Frid MG, Flockton AR, McKeon BA, Yeager ME, Fini MA, Morrell NW, Pullamsetti SS, Velegala S, Seeger W, McKinsey TA, Sucharov CC, Stenmark KR. MicroRNA-124 controls the proliferative, migratory, and inflammatory phenotype of pulmonary vascular fibroblasts. Circ Res 2013; 114:67-78. [PMID: 24122720 DOI: 10.1161/circresaha.114.301633] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
RATIONALE Pulmonary hypertensive remodeling is characterized by excessive proliferation, migration, and proinflammatory activation of adventitial fibroblasts. In culture, fibroblasts maintain a similar activated phenotype. The mechanisms responsible for generation/maintenance of this phenotype remain unknown. OBJECTIVE We hypothesized that aberrant expression of microRNA-124 (miR-124) regulates this activated fibroblast phenotype and sought to determine the signaling pathways through which miR-124 exerts effects. METHODS AND RESULTS We detected significant decreases in miR-124 expression in fibroblasts isolated from calves and humans with severe pulmonary hypertension. Overexpression of miR-124 by mimic transfection significantly attenuated proliferation, migration, and monocyte chemotactic protein-1 expression of hypertensive fibroblasts, whereas anti-miR-124 treatment of control fibroblasts resulted in their increased proliferation, migration, and monocyte chemotactic protein-1 expression. Furthermore, the alternative splicing factor, polypyrimidine tract-binding protein 1, was shown to be a direct target of miR-124 and to be upregulated both in vivo and in vitro in bovine and human pulmonary hypertensive fibroblasts. The effects of miR-124 on fibroblast proliferation were mediated via direct binding to the 3' untranslated region of polypyrimidine tract-binding protein 1 and subsequent regulation of Notch1/phosphatase and tensin homolog/FOXO3/p21Cip1 and p27Kip1 signaling. We showed that miR-124 directly regulates monocyte chemotactic protein-1 expression in pulmonary hypertension/idiopathic pulmonary arterial hypertension fibroblasts. Furthermore, we demonstrated that miR-124 expression is suppressed by histone deacetylases and that treatment of hypertensive fibroblasts with histone deacetylase inhibitors increased miR-124 expression and decreased proliferation and monocyte chemotactic protein-1 production. CONCLUSIONS Stable decreases in miR-124 expression contribute to an epigenetically reprogrammed, highly proliferative, migratory, and inflammatory phenotype of hypertensive pulmonary adventitial fibroblasts. Thus, therapies directed at restoring miR-124 function, including histone deacetylase inhibitors, should be investigated.
Collapse
Affiliation(s)
- Daren Wang
- From the Department of Pediatrics (D.W., H.Z., M.L., M.G.F., A.R.F., B.A.K., M.E.Y., M.A.F.), Department of Medicine (T.A.M., C.C.S.), Department of Medicine and Pediatrics (K.R.S.), Department of Medicine (N.W.M.), Department of Lung Development and Remodeling (S.S.P., S.V., W.S.), Department of Medicine (H.Z.), University of Colorado Anschutz Medical Campus, Aurora, CO; University of Cambridge, Cambridge, United Kingdom (N.W.M.); Addenbrooke's & Papworth Hospitals, Cambridge, United Kingdom (N.W.M.); Max-Planck-Institute for Heart and Lung Research; University of Giessen and Marburg Lung Center, Bad Nauheim, Germany (S.S.P., S.V., W.S.); and Shengjing Hospital of China Medical University, Shenyang, China (H.Z.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Neuman MG, Nanau RM, Shekh-Ahmad T, Yagen B, Bialer M. Valproic acid derivatives signal for apoptosis and repair in vitro. Clin Biochem 2013; 46:1532-7. [DOI: 10.1016/j.clinbiochem.2013.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 06/17/2013] [Accepted: 07/02/2013] [Indexed: 11/26/2022]
|
186
|
The HDAC inhibitor LBH589 induces ERK-dependent prometaphase arrest in prostate cancer via HDAC6 inactivation and down-regulation. PLoS One 2013; 8:e73401. [PMID: 24023871 PMCID: PMC3762759 DOI: 10.1371/journal.pone.0073401] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 07/19/2013] [Indexed: 01/03/2023] Open
Abstract
Histone deacetylase inhibitors (HDACIs) have potent anti-cancer activity in a variety of cancer models. Understanding the molecular mechanisms involved in the therapeutic responsiveness of HDACI is needed before its clinical application. This study aimed to determine if a potent HDACI, LBH589 (Panobinostat), had differential therapeutic responsiveness towards LNCaP and PC-3 prostate cancer (PCa) cells. The former showed prometaphase arrest with subsequent apoptosis upon LBH589 treatment, while the latter was less sensitive and had late G2 arrest. The LBH589 treatment down-regulated HDAC6 and sustained ERK activation, and contributed to prometaphase arrest. Mechanistically, LBH589 inhibited HDAC6 activity, caused its dissociation from protein phosphatase PP1α, and increased 14-3-3ζ acetylation. Acetylated 14-3-3ζ released its mask effect on serine 259 of c-Raf and serine 216 of Cdc25C subsequent to de-phosphorylation by PP1α, which contributed to ERK activation. Enhanced ERK activity by LBH589 further down-regulated HDAC6 protein levels and sustained ERK activation by free-forward regulation. The sustained Cdc25C and ERK activation resulted in early M-phase (prometaphase) arrest and subsequent apoptosis in the most sensitive LNCaP cells but not in PC-3 cells. This study provides pre-clinical evidence that HDAC6 may serve as a sensitive therapeutic target in the treatment of prostate cancer with HDACI LBH589 for clinical translation. This study also posits a novel mechanism of HDAC6 participation in regulating the c-Raf-PP1-ERK signaling pathway and contributing to M phase cell-cycle transition.
Collapse
|
187
|
Giaginis C, Alexandrou P, Delladetsima I, Giannopoulou I, Patsouris E, Theocharis S. Clinical significance of histone deacetylase (HDAC)-1, HDAC-2, HDAC-4, and HDAC-6 expression in human malignant and benign thyroid lesions. Tumour Biol 2013; 35:61-71. [PMID: 23873102 DOI: 10.1007/s13277-013-1007-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 07/05/2013] [Indexed: 01/08/2023] Open
Abstract
Histone deacetylases (HDACs) have been associated with human malignant tumor development and progression, and HDAC inhibitors are currently being explored as anticancer agents in clinical trials. The present study aimed to evaluate the clinical significance of HDAC-1, HDAC-2, HDAC-4, and HDAC-6 proteins' expression in human malignant and benign thyroid lesions. HDAC-1, HDAC-2, HDAC-4, and HDAC-6 proteins' expression was assessed immunohistochemically on paraffin-embedded thyroid tissues obtained from 74 patients with benign and malignant thyroid lesions. Enhanced HDAC-2 and HDAC-6 expression was significantly more frequently observed in malignant, compared to benign, thyroid lesions (p = 0.0042 and p = 0.0069, respectively). Enhanced HDAC-2, HDAC-4, and HDAC-6 expression was significantly more frequently observed in cases with papillary carcinoma compared to hyperplastic nodules (p = 0.0065, p = 0.0394, and p = 0.0061, respectively). In malignant thyroid lesions, HDAC-1, HDAC-4, and HDAC-6 expression was significantly associated with tumor size (p = 0.0169, p = 0.0056, and p = 0.0234, respectively); HDAC-2 expression with lymphatic and vascular invasion (p = 0.0299 and p = 0.0391, respectively); and HDAC-4 expression with capsular invasion (p = 0.0464). The cellular pattern of HDAC-1 and HDAC-2 distribution (nuclear vs. nuclear and cytoplasmic) presented a distinct discrimination between malignant and benign thyroid lesions (p = 0.0030 and p = 0.0028, respectively) as well as between papillary carcinoma and hyperplastic nodules (p = 0.0036 and p = 0.0028, respectively). HDAC-1, HDAC-2, HDAC-4, and HDAC-6 may be associated with the malignant thyroid transformation and could be considered as useful biomarkers and possible therapeutic targets in this neoplasia.
Collapse
Affiliation(s)
- Constantinos Giaginis
- First Department of Pathology, Medical School, University of Athens, 75 M. Asias str., Goudi, Athens, GR11527, Greece
| | | | | | | | | | | |
Collapse
|
188
|
Abstract
BAP1 (BRCA1-Associated Protein 1) was initially identified as a protein that binds to BRCA1. BAP1 is a tumour suppressor that is believed to mediate its effects through chromatin modulation, transcriptional regulation, and possibly via the ubiquitin-proteasome system and the DNA damage response pathway. Germline mutations of BAP1 confer increased susceptibility for the development of several tumours, including uveal melanoma, epithelioid atypical Spitz tumours, cutaneous melanoma, and mesothelioma. However, the complete tumour spectrum associated with germline BAP1 mutations is not yet known. Somatic BAP1 mutations are seen in cutaneous melanocytic tumours (epithelioid atypical Spitz tumours and melanoma), uveal melanoma, mesothelioma, clear cell renal cell carcinoma, and other tumours. Here, we review the current state of knowledge about the functional roles of BAP1, and summarise data on tumours associated with BAP1 mutations. Awareness of these tumours will help pathologists and clinicians to identify patients with a high likelihood of harbouring germline or somatic BAP1 mutations. We recommend that pathologists consider testing for BAP1 mutations in epithelioid atypical Spitz tumours and uveal melanomas, or when other BAP1-associated tumours occur in individual patients. Tumour tissues may be screened for BAP1 mutations/loss/inactivation by immunohistochemistry (IHC) (demonstrated by loss of nuclear staining in tumour cells). Confirmatory sequencing may be considered in tumours that exhibit BAP1 loss by IHC and in those with equivocal IHC results. If a BAP1 mutation is confirmed in a tumour, the patient's treating physician should be informed of the possibility of a BAP1 germline mutation, so they can consider whether genetic counselling and further testing of the patient and investigation of their family is appropriate. Recognition and evaluation of larger numbers of BAP1-associated tumours will also be necessary to facilitate identification of additional distinct clinico-pathological characteristics or other genotype-phenotype correlations that may have prognostic and management implications.
Collapse
|
189
|
Transcriptional control of cancer metastasis. Trends Cell Biol 2013; 23:603-11. [PMID: 23838335 DOI: 10.1016/j.tcb.2013.06.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 06/05/2013] [Accepted: 06/06/2013] [Indexed: 12/13/2022]
Abstract
Transcriptional regulation is an essential component of tumor progression and metastasis. During cancer progression, dysregulation of oncogenic or tumor-suppressive transcription factors (TFs), as well as master cell fate regulators and tumor microenvironment-induced factors, collectively influence multiple steps of the metastasis cascade, including local invasion, dissemination, and eventual colonization of the tumor to distant organs. Furthermore, epigenetic alterations in tumor cells, including DNA methylation, as well as activation or suppression of histone deacetylases (HDACs), histone acetyltransferases (HATs), and other chromatin-modifying enzymes, can further distort the transcriptional network to influence metastasis. We focus here on recent research advances in transcriptional control of metastasis and highlight the therapeutic potential of targeting such transcriptional regulatory networks.
Collapse
|
190
|
André N, Banavali S, Snihur Y, Pasquier E. Has the time come for metronomics in low-income and middle-income countries? Lancet Oncol 2013; 14:e239-48. [PMID: 23639324 DOI: 10.1016/s1470-2045(13)70056-1] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In 2008, 72% of cancer deaths occurred in low-income and middle-income countries, where, although there is a lower incidence of cancer than in high-income countries, survival rates are also low. Many patients are sent home to die, and an even larger number of patients do not have access to treatment facilities. New constraint-adapted therapeutic strategies are therefore urgently needed. Metronomic chemotherapy--the chronic administration of chemotherapy at low, minimally toxic doses on a frequent schedule of administration, with no prolonged drug-free breaks--has recently emerged as a potential strategy to control advanced or refractory cancer and represents an alternative for patients with cancer living in developing countries. This low-cost, well-tolerated, and easy to access strategy is an attractive therapeutic option in resource-limited countries. Moreover, combined with drug repositioning, additional anticancer effects can be achieved, ultimately resulting in improved cancer control while maintaining minimum cost of treatment. In this Personal View, we will briefly review the rationale behind the combination of metronomic chemotherapy and drug repositioning-an approach we term metronomics. We assess the clinical experience obtained with this kind of anticancer treatment and describe potential new developments in countries with limited resources. We also highlight the need for adapted clinical study endpoints and innovative models of collaboration between for-profit and non-profit organisations, to address the growing problem of cancer in resource-limited countries.
Collapse
Affiliation(s)
- Nicolas André
- Service d'Hématologie et Oncologie Pédiatrique, AP-HM, Marseille, France.
| | | | | | | |
Collapse
|
191
|
Van Lint C, Bouchat S, Marcello A. HIV-1 transcription and latency: an update. Retrovirology 2013; 10:67. [PMID: 23803414 PMCID: PMC3699421 DOI: 10.1186/1742-4690-10-67] [Citation(s) in RCA: 247] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 05/29/2013] [Indexed: 12/11/2022] Open
Abstract
Combination antiretroviral therapy, despite being potent and life-prolonging, is not curative and does not eradicate HIV-1 infection since interruption of treatment inevitably results in a rapid rebound of viremia. Reactivation of latently infected cells harboring transcriptionally silent but replication-competent proviruses is a potential source of persistent residual viremia in cART-treated patients. Although multiple reservoirs may exist, the persistence of resting CD4+ T cells carrying a latent infection represents a major barrier to eradication. In this review, we will discuss the latest reports on the molecular mechanisms that may regulate HIV-1 latency at the transcriptional level, including transcriptional interference, the role of cellular factors, chromatin organization and epigenetic modifications, the viral Tat trans-activator and its cellular cofactors. Since latency mechanisms may also operate at the post-transcriptional level, we will consider inhibition of nuclear RNA export and inhibition of translation by microRNAs as potential barriers to HIV-1 gene expression. Finally, we will review the therapeutic approaches and clinical studies aimed at achieving either a sterilizing cure or a functional cure of HIV-1 infection, with a special emphasis on the most recent pharmacological strategies to reactivate the latent viruses and decrease the pool of viral reservoirs.
Collapse
Affiliation(s)
- Carine Van Lint
- Université Libre de Bruxelles (ULB), Service of Molecular Virology, Institute of Molecular Biology and Medicine, 12, Rue des Profs Jeener et Brachet, 6041, Gosselies, Belgium.
| | | | | |
Collapse
|
192
|
Nanau RM, Neuman MG. Adverse drug reactions induced by valproic acid. Clin Biochem 2013; 46:1323-38. [PMID: 23792104 DOI: 10.1016/j.clinbiochem.2013.06.012] [Citation(s) in RCA: 193] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 06/11/2013] [Accepted: 06/12/2013] [Indexed: 12/11/2022]
Abstract
Valproic acid is a widely-used first-generation antiepileptic drug, prescribed predominantly in epilepsy and psychiatric disorders. VPA has good efficacy and pharmacoeconomic profiles, as well as a relatively favorable safety profile. However, adverse drug reactions have been reported in relation with valproic acid use, either as monotherapy or polytherapy with other antiepileptic drugs or antipsychotic drugs. This systematic review discusses valproic acid adverse drug reactions, in terms of hepatotoxicity, mitochondrial toxicity, hyperammonemic encephalopathy, hypersensitivity syndrome reactions, neurological toxicity, metabolic and endocrine adverse events, and teratogenicity.
Collapse
Affiliation(s)
- Radu M Nanau
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, In Vitro Drug Safety and Biotechnology, Toronto, Ontario, Canada
| | | |
Collapse
|
193
|
Fan J, Alsarraf O, Dahrouj M, Platt KA, Chou CJ, Rice DS, Crosson CE. Inhibition of HDAC2 protects the retina from ischemic injury. Invest Ophthalmol Vis Sci 2013; 54:4072-80. [PMID: 23696608 DOI: 10.1167/iovs.12-11529] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
PURPOSE Protein acetylation is an essential mechanism in regulating transcriptional and inflammatory events. Studies have shown that nonselective histone deacetylase (HDAC) inhibitors can protect the retina from ischemic injury in rats. However, the role of specific HDAC isoforms in retinal degenerative processes remains obscure. The purpose of this study was to investigate the role of HDAC2 isoform in a mouse model of ischemic retinal injury. METHODS Localization of HDAC2 in mice retinas was evaluated by immunohistochemical analyses. To investigate whether selective reduction in HDAC2 activity can protect the retina from ischemic injury, Hdac2⁺/⁻ mice were utilized. Electroretinographic (ERG) and morphometric analyses were used to assess retinal function and morphology. RESULTS Our results demonstrated that HDAC2 is primarily localized in nuclei in inner nuclear and retinal ganglion cell layers, and HDAC2 activity accounted for approximately 35% of the total activities of HDAC1, 2, 3, and 6 in the retina. In wild-type mice, ERG a- and b-waves from ischemic eyes were significantly reduced when compared to pre-ischemia baseline values. Morphometric examination of these eyes revealed significant degeneration of inner retinal layers. In Hdac2⁺/⁻ mice, ERG a- and b-waves from ischemic eyes were significantly greater than those measured in ischemic eyes from wild-type mice. Morphologic measurements demonstrated that Hdac2⁺/⁻ mice exhibit significantly less retinal degeneration than wild-type mice. CONCLUSIONS This study demonstrated that suppressing HDAC2 expression can effectively reduce ischemic retinal injury. Our results support the idea that the development of selective HDAC2 inhibitors may provide an efficacious treatment for ischemic retinal injury.
Collapse
Affiliation(s)
- Jie Fan
- Storm Eye Institute, Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina 29425, USA.
| | | | | | | | | | | | | |
Collapse
|
194
|
Summers AR, Fischer MA, Stengel KR, Zhao Y, Kaiser JF, Wells CE, Hunt A, Bhaskara S, Luzwick JW, Sampathi S, Chen X, Thompson MA, Cortez D, Hiebert SW. HDAC3 is essential for DNA replication in hematopoietic progenitor cells. J Clin Invest 2013; 123:3112-23. [PMID: 23921131 DOI: 10.1172/jci60806] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 04/16/2013] [Indexed: 11/17/2022] Open
Abstract
Histone deacetylase 3 (HDAC3) contributes to the regulation of gene expression, chromatin structure, and genomic stability. Because HDAC3 associates with oncoproteins that drive leukemia and lymphoma, we engineered a conditional deletion allele in mice to explore the physiological roles of Hdac3 in hematopoiesis. We used the Vav-Cre transgenic allele to trigger recombination, which yielded a dramatic loss of lymphoid cells, hypocellular bone marrow, and mild anemia. Phenotypic and functional analysis suggested that Hdac3 was required for the formation of the earliest lymphoid progenitor cells in the marrow, but that the marrow contained 3-5 times more multipotent progenitor cells. Hdac3(-/-) stem cells were severely compromised in competitive bone marrow transplantation. In vitro, Hdac3(-/-) stem and progenitor cells failed to proliferate, and most cells remained undifferentiated. Moreover, one-third of the Hdac3(-/-) stem and progenitor cells were in S phase 2 hours after BrdU labeling in vivo, suggesting that these cells were impaired in transit through the S phase. DNA fiber-labeling experiments indicated that Hdac3 was required for efficient DNA replication in hematopoietic stem and progenitor cells. Thus, Hdac3 is required for the passage of hematopoietic stem/progenitor cells through the S phase, for stem cell functions, and for lymphopoiesis.
Collapse
Affiliation(s)
- Alyssa R Summers
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
195
|
Choudhary S, Sood S, Wang HCR. Synergistic induction of cancer cell death and reduction of clonogenic resistance by cisplatin and FK228. Biochem Biophys Res Commun 2013; 436:325-30. [PMID: 23743194 DOI: 10.1016/j.bbrc.2013.05.102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 05/23/2013] [Indexed: 01/29/2023]
Abstract
Human urinary bladder cancer is the fifth most common cancer in the United States, and the long-term disease-free survival in patients is still suboptimal with current chemotherapeutic regimens. Development of effective chemotherapeutic regimens is crucial to decrease the morbidity and mortality of this cancer. The goal of this study was to investigate the effectiveness of FK228 in increasing cisplatin's ability to induce bladder cancer cell death and reduce drug resistance. Our study revealed that FK228 combined with cisplatin synergistically induced cell death and reduced clonogenic survival of human urinary bladder cancer cells. The Erk-Nox pathway played an important role in mediating signals highly increased by this combined treatment to induce significantly-elevated levels of reactive oxygen species, leading to substantially-induced caspase activation and synergistically-increased death in cancer cells. Cisplatin was able to enhance the ability of FK228 to significantly reduce glutathione, indicating a novel activity of combined FK228 and cisplatin in reducing drug resistance. The ability of combined FK228 and cisplatin to synergistically induce cell death and reduce clonogenic survival was also applicable to colon cancer cells. Hence, combined use of FK228 with cisplatin should be considered in development of therapeutic strategies to control urinary bladder cancer and other cancer development and recurrence.
Collapse
Affiliation(s)
- Shambhunath Choudhary
- Anticancer Molecular Oncology Laboratory, Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN 37996, United States
| | | | | |
Collapse
|
196
|
Guo SQ, Zhang YZ. Histone deacetylase inhibition: an important mechanism in the treatment of lymphoma. Cancer Biol Med 2013; 9:85-9. [PMID: 23691460 PMCID: PMC3643654 DOI: 10.3969/j.issn.2095-3941.2012.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 06/13/2012] [Indexed: 12/16/2022] Open
Abstract
Lymphomas encompass a group of malignancies that originate in the lymph nodes or other lymphoid tissues. Epigenetic modification, especially by histone deacetylase (HDACs), plays a key role during the occurrence and development of lymphomas. Consequently, HDAC inhibitors (HDACIs), a class of gene expression-modulating drugs, have emerged as promising mechanism-based agents for the treatment of lymphomas. This review presents the rationale of HDAC inhibition, describes the epigenetic-based mechanisms of action of HDACIs, discusses their clinical efficiency, and summarizes the current and future developments in this field.
Collapse
Affiliation(s)
- Shan-Qi Guo
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin 300060, China
| | | |
Collapse
|
197
|
Kerkhof M, Dielemans JCM, van Breemen MS, Zwinkels H, Walchenbach R, Taphoorn MJ, Vecht CJ. Effect of valproic acid on seizure control and on survival in patients with glioblastoma multiforme. Neuro Oncol 2013; 15:961-7. [PMID: 23680820 DOI: 10.1093/neuonc/not057] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND To examine the efficacy of valproic acid (VPA) given either with or without levetiracetam (LEV) on seizure control and on survival in patients with glioblastoma multiforme (GBM) treated with chemoradiation. METHODS A retrospective analysis was performed on 291 patients with GBM. The efficacies of VPA and LEV alone and as polytherapy were analyzed in 181 (62%) patients with seizures with a minimum follow-up of 6 months. Cox-regression survival analysis was performed on 165 patients receiving chemoradiation with temozolomide of whom 108 receiving this in combination with VPA for at least 3 months. RESULTS Monotherapy with either VPA or LEV was instituted in 137/143 (95.8%) and in 59/86 (68.6%) on VPA/LEV polytherapy as the next regimen. Initial freedom from seizure was achieved in 41/100 (41%) on VPA, in 16/37 (43.3%) on LEV, and in 89/116 (76.7%) on subsequent VPA/LEV polytherapy. At the end of follow-up, seizure freedom was achieved in 77.8% (28/36) on VPA alone, in 25/36 (69.5%) on LEV alone, and in 38/63 (60.3%) on VPA/LEV polytherapy with ongoing seizures on monotherapy. Patients using VPA in combination with temozolomide showed a longer median survival of 69 weeks (95% confidence interval [CI]: 61.7-67.3) compared with 61 weeks (95% CI: 52.5-69.5) in the group without VPA (hazard ratio, 0.63; 95% CI: 0.43-0.92; P = .016), adjusting for age, extent of resection, and O(6)-DNA methylguanine-methyltransferase promoter methylation status. CONCLUSIONS Polytherapy with VPA and LEV more strongly contributes to seizure control than does either as monotherapy. Use of VPA together with chemoradiation with temozolomide results in a 2-months' longer survival of patients with GBM.
Collapse
Affiliation(s)
- Melissa Kerkhof
- Neuro-oncology Unit, Department of Neurology,Medical Center Haaglanden, The Hague, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
198
|
Xu W, Li Y, Liu C, Zhao S. Protein lysine acetylation guards metabolic homeostasis to fight against cancer. Oncogene 2013; 33:2279-85. [PMID: 23665675 DOI: 10.1038/onc.2013.163] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 03/11/2013] [Accepted: 03/12/2013] [Indexed: 02/07/2023]
Abstract
Properly coordinated metabolism and maintained metabolite homeostasis are important because altered metabolite homeostasis has a causal role in many human diseases, including cancer. Metabolite homeostasis is maintained by fine-tuned coordination of metabolite generation and utilization. Metabolite deregulation has recently been shown to alter the signaling pathways and reprogram epigenetic factors associated with tumorigenesis. Protein lysine acetylation is emerging as a metabolism-coordinating mechanism. Mechanistic studies have shown that acetylation may have roles in nutrient adaptation and in maintaining metabolite homeostasis by exerting regulatory effects on metabolic enzymes, metabolic pathways and metabolic networks. Here we review recent progress in the determination of the role of acetylation regulation in metabolism coordination. In particular, we review links between deregulated acetylation in metabolic enzymes and tumorigenesis. We further hypothesize on applications of the mediation of acetylation to restore deregulated metabolism coordination and thus develop novel means of cancer treatment.
Collapse
Affiliation(s)
- W Xu
- 1] Sate Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People's Republic of China [2] Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Y Li
- Sate Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People's Republic of China
| | - C Liu
- Sate Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People's Republic of China
| | - S Zhao
- 1] Sate Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People's Republic of China [2] Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
199
|
Zdrenghea MT. Could interleukin-15 potentiate histone deacetylase inhibitor effects in haematological malignancy? Med Hypotheses 2013; 81:311-5. [PMID: 23669372 DOI: 10.1016/j.mehy.2013.04.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 03/29/2013] [Accepted: 04/14/2013] [Indexed: 01/10/2023]
Abstract
Despite significant progress in cancer therapy, prognosis in acute leukaemia remains dismal, and the development of new therapies is urgently warranted: in acute myeloid leukaemia, the current cure rate is of 30-40% in younger and much less in older patients. Chromatin remodeling through histone acetylation is one of the major mechanisms of transcriptional control of genes, and is involved in 'gene silencing' of antioncogenes in various tumour cells. Chromatin remodeling is also involved in transcriptional control of other genes, such as NKG2D ligand genes. Histone deacetylases and acetyltransferases are involved in the epigenetic regulation of gene expression, and increased/decreased activity of histone deacetylases has been reported in several cancer types. Histone deacetylase inhibitors were reportedly active in many cancers including hematological malignancies, and have been shown in numerous experiments to reduce cancer cell growth and enhance cell differentiation, growth arrest and apoptosis. In acute myeloid leukaemia, histone deacetylase inhibitors alone had limited efficacy, but their combination with other anticancer agents yielded promising results. Interleukin (IL)-15 is regarded with great hope in the immunotherapy of cancer, and IL-15-activated cytokine-induced killer cells showed potent antileukemic activity both in vitro and in vivo. IL-15 increases expression of NKG2D and its ligands and can increase natural killer cell mediated cytotoxicity against tumour cells. The administration of IL-15 was recently shown to be safe in preclinical models, and there are ongoing clinical trials of IL-15 in patients with cancer and HIV infection. We hypothesise that IL-15 will synergise with histone deacetylase inhibitors in increasing the levels of activatory NKG2D receptors on natural killer and CD8(+) T cells and of their ligands, the MHC class I related molecule A and B, on tumor cells, and will enhance innate immune antitumour responses in acute myeloid leukaemia and other haematological malignancies. Up-regulation of NKG2D-NKG2D-ligand antitumour immune response by combining histone deacetylase inhibitors with IL-15 has the potential to improve the efficacy of acute myeloid leukaemia treatment.
Collapse
Affiliation(s)
- Mihnea T Zdrenghea
- Oncology Institute, University of Medicine and Pharmacy Cluj-Napoca 73, 21 December Blvd, 400124 Cluj-Napoca, Romania.
| |
Collapse
|
200
|
Ossenkoppele GJ, Lowenberg B, Zachee P, Vey N, Breems D, Van de Loosdrecht AA, Davidson AH, Wells G, Needham L, Bawden L, Toal M, Hooftman L, Debnam PM. A phase I first-in-human study with tefinostat - a monocyte/macrophage targeted histone deacetylase inhibitor - in patients with advanced haematological malignancies. Br J Haematol 2013; 162:191-201. [PMID: 23647373 DOI: 10.1111/bjh.12359] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 01/25/2013] [Indexed: 12/26/2022]
Abstract
Tefinostat (CHR-2845) is a monocyte/macrophage targeted histone deacetylase inhibitor (HDACi). This first-in-human, standard 3 + 3 dose escalating trial of oral, once daily tefinostat was conducted to determine the safety, tolerability, pharmacokinetic and pharmacodynamic profile of tefinostat in relapsed/refractory haematological diseases. Eighteen patients were enrolled at doses of 20-640 mg. Plasma concentrations of tefinostat exceeded those demonstrated to give in vitro anti-proliferative activity. Flow cytometric pharmacodynamic assays demonstrated monocyte-targeted increases in protein acetylation, without corresponding changes in lymphocytes. Dose-limiting toxicities (DLTs) were not observed and dose escalation was halted at 640 mg without identification of the maximum tolerated dose. Drug-related toxicities were largely Common Toxicity Criteria for Adverse Events grade 1/2 and included nausea, anorexia, fatigue, constipation, rash and increased blood creatinine. A patient with chronic monomyelocytic leukaemia achieved a bone marrow response, with no change in peripheral monocytes. An acute myeloid leukaemia type M2 patient showed a >50% decrease in bone marrow blasts and clearance of peripheral blasts. In conclusion, tefinostat produces monocyte-targeted HDACi activity and is well tolerated, without the DLTs, e.g. fatigue, diarrhoea, thrombocytopenia, commonly seen with non-targeted HDACi. The early signs of efficacy and absence of significant toxicity warrant further evaluation of tefinostat in larger studies. (clinicaltrials.gov identifier: NCT00820508).
Collapse
Affiliation(s)
- Gert J Ossenkoppele
- Department of Haematology, VU University Medical Centre, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|