1
|
Singh D, Gupta S. Butyrate: A Review on Beneficial Pharmacological and Therapeutic Effect. CURRENT NUTRITION & FOOD SCIENCE 2021. [DOI: 10.2174/1573401316999201029210912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background::
Short-chain fatty acids (SCFAs), generally acetate, propionate along with
butyrate, are aliphatic organic acids formed in the gut mucosa through bacterial fermentation of
mostly undigested nutritional carbohydrates, again to a minor degree by natural and dietary proteins,
such as mucous and shed epithelial cells.
Methods::
Many sources were used to collect information about Butyrate, such as Pub med, Google
Scholar, Pubmed, Scopus and other reliable sources.
:
Endogenous butyrate formation, absorption, and transportation by colon cells have now been well
acknowledged. Butyrate exerts its action features by way of appearing as a histone deacetylase inhibitor,
even signaling through a few protein receptors. Lately, butyrate has received special consideration
for its favorable result on intestinal equilibrium and also energy metabolism. There is a
growing interest in butyrate as its impact on epigenetic mechanisms will result in much more certain
and also efficacious healing techniques for the prevention and therapy of various diseases that
range from genetic conditions to other body disorders.
Conclusion::
With this assessment, we compile the existing information on the attributes of butyrate,
particularly its potential effects and also mechanisms involved in cancer, inflammation, diabetes
mellitus, neurological and cardiovascular disorder.
Collapse
Affiliation(s)
- Dhirendra Singh
- Department of Pharmacology, M.M. College of Pharmacy, (Deemed to be University), Mullana, Ambala, Haryana, India
| | - Sumeet Gupta
- Department of Pharmacology, M.M. College of Pharmacy, (Deemed to be University), Mullana, Ambala, Haryana, India
| |
Collapse
|
2
|
Ramaiah MJ, Tangutur AD, Manyam RR. Epigenetic modulation and understanding of HDAC inhibitors in cancer therapy. Life Sci 2021; 277:119504. [PMID: 33872660 DOI: 10.1016/j.lfs.2021.119504] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/20/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022]
Abstract
The role of genetic and epigenetic factors in tumor initiation and progression is well documented. Histone deacetylases (HDACs), histone methyl transferases (HMTs), and DNA methyl transferases. (DNMTs) are the main proteins that are involved in regulating the chromatin conformation. Among these, histone deacetylases (HDAC) deacetylate the histone and induce gene repression thereby leading to cancer. In contrast, histone acetyl transferases (HATs) that include GCN5, p300/CBP, PCAF, Tip 60 acetylate the histones. HDAC inhibitors are potent drug molecules that can induce acetylation of histones at lysine residues and induce open chromatin conformation at tumor suppressor gene loci and thus resulting in tumor suppression. The key processes regulated by HDAC inhibitors include cell-cycle arrest, chemo-sensitization, apoptosis induction, upregulation of tumor suppressors. Even though FDA approved drugs are confined mainly to haematological malignancies, the research on HDAC inhibitors in glioblastoma multiforme and triple negative breast cancer (TNBC) are providing positive results. Thus, several combinations of HDAC inhibitors along with DNA methyl transferase inhibitors and histone methyl transferase inhibitors are in clinical trials. This review focuses on how HDAC inhibitors regulate the expression of coding and non-coding genes with specific emphasis on their anti-cancer potential.
Collapse
Affiliation(s)
- M Janaki Ramaiah
- Laboratory of Functional genomics and Disease Biology, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, Tamil Nadu, India.
| | - Anjana Devi Tangutur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500 007, Telangana, India
| | - Rajasekhar Reddy Manyam
- Department of Computer Science and Engineering, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Andhra Pradesh, India
| |
Collapse
|
3
|
Mahmud SMH, Al-Mustanjid M, Akter F, Rahman MS, Ahmed K, Rahman MH, Chen W, Moni MA. Bioinformatics and system biology approach to identify the influences of SARS-CoV-2 infections to idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease patients. Brief Bioinform 2021; 22:6224261. [PMID: 33847347 PMCID: PMC8083324 DOI: 10.1093/bib/bbab115] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/25/2021] [Accepted: 03/13/2021] [Indexed: 12/15/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), better known as COVID-19, has become a current threat to humanity. The second wave of the SARS-CoV-2 virus has hit many countries, and the confirmed COVID-19 cases are quickly spreading. Therefore, the epidemic is still passing the terrible stage. Having idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD) are the risk factors of the COVID-19, but the molecular mechanisms that underlie IPF, COPD, and CVOID-19 are not well understood. Therefore, we implemented transcriptomic analysis to detect common pathways and molecular biomarkers in IPF, COPD, and COVID-19 that help understand the linkage of SARS-CoV-2 to the IPF and COPD patients. Here, three RNA-seq datasets (GSE147507, GSE52463, and GSE57148) from Gene Expression Omnibus (GEO) is employed to detect mutual differentially expressed genes (DEGs) for IPF, and COPD patients with the COVID-19 infection for finding shared pathways and candidate drugs. A total of 65 common DEGs among these three datasets were identified. Various combinatorial statistical methods and bioinformatics tools were used to build the protein–protein interaction (PPI) and then identified Hub genes and essential modules from this PPI network. Moreover, we performed functional analysis under ontologies terms and pathway analysis and found that IPF and COPD have some shared links to the progression of COVID-19 infection. Transcription factors–genes interaction, protein–drug interactions, and DEGs-miRNAs coregulatory network with common DEGs also identified on the datasets. We think that the candidate drugs obtained by this study might be helpful for effective therapeutic in COVID-19.
Collapse
Affiliation(s)
- S M Hasan Mahmud
- Computer Science and Technology from the University of Electronic Science and Technology of China, China
| | | | - Farzana Akter
- Computer Science and Engineering from Daffodil International University, Bangladesh
| | | | - Kawsar Ahmed
- Information and Communication Technology (ICT) at Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Md Habibur Rahman
- Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Wenyu Chen
- University of Electronic Science and Technology of China, China
| | | |
Collapse
|
4
|
Cao T, Zhang X, Chen D, Zhang P, Li Q, Muhammad A. The epigenetic modification during the induction of Foxp3 with sodium butyrate. Immunopharmacol Immunotoxicol 2018; 40:309-318. [DOI: 10.1080/08923973.2018.1480631] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Tengli Cao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiuxiu Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Dingding Chen
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Peiyan Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qing Li
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Abbas Muhammad
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
5
|
Kovacs L, Kovacs-Kasa A, Verin AD, Fulton D, Lucas R, Su Y. Histone deacetylases in vascular permeability and remodeling associated with acute lung injury. ACTA ACUST UNITED AC 2018; 2. [PMID: 32099966 DOI: 10.20517/2574-1209.2018.06] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Acute lung injury (ALI) is a severe progressive disorder that arises from a wide range of causes such as toxins or inflammation, resulting in significant morbidity and mortality. There are no effective therapeutic options apart from mechanical ventilation strategies. While the mechanisms that govern the clinically relevant process of increased EC permeability and remodeling associated with ALI are under intense investigation, our knowledge of the processes that determine barrier enhancement or preservation are far from completion. Recently, epigenetic mechanisms have emerged as a major regulator of enduring changes in cell behavior and the therapeutic potential of inhibiting histone deacetylases (HDACs) for the treatment of cardiovascular and inflammatory diseases has gained remarkable attention. Although HDACs have been shown to play an important role in regulating EC barrier function, the involved HDAC subtypes and mechanisms remain undefined. Further investigation of the HDAC signaling may provide therapeutic approaches for the prevention and treatment of ALI.
Collapse
Affiliation(s)
- Laszlo Kovacs
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA 30912
| | | | - Alexander D Verin
- Vascular Biology Center, Augusta University, Augusta, GA 30912.,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - David Fulton
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA 30912.,Vascular Biology Center, Augusta University, Augusta, GA 30912
| | - Rudolf Lucas
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA 30912.,Vascular Biology Center, Augusta University, Augusta, GA 30912.,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Yunchao Su
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA 30912.,Vascular Biology Center, Augusta University, Augusta, GA 30912.,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912.,Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia 30912
| |
Collapse
|
6
|
Di Martile M, Desideri M, De Luca T, Gabellini C, Buglioni S, Eramo A, Sette G, Milella M, Rotili D, Mai A, Carradori S, Secci D, De Maria R, Del Bufalo D, Trisciuoglio D. Histone acetyltransferase inhibitor CPTH6 preferentially targets lung cancer stem-like cells. Oncotarget 2017; 7:11332-48. [PMID: 26870991 PMCID: PMC4905477 DOI: 10.18632/oncotarget.7238] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/23/2016] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) play an important role in tumor initiation, progression, therapeutic failure and tumor relapse. In this study, we evaluated the efficacy of the thiazole derivative 3-methylcyclopentylidene-[4-(4′-chlorophenyl)thiazol-2-yl]hydrazone (CPTH6), a novel pCAF and Gcn5 histone acetyltransferase inhibitor, as a small molecule that preferentially targets lung cancer stem-like cells (LCSCs) derived from non-small cell lung cancer (NSCLC) patients. Notably, although CPTH6 inhibits the growth of both LCSC and NSCLC cell lines, LCSCs exhibit greater growth inhibition than established NSCLC cells. Growth inhibitory effect of CPTH6 in LCSC lines is primarily due to apoptosis induction. Of note, differentiated progeny of LCSC lines is more resistant to CPTH6 in terms of loss of cell viability and reduction of protein acetylation, when compared to their undifferentiated counterparts. Interestingly, in LCSC lines CPTH6 treatment is also associated with a reduction of stemness markers. By using different HAT inhibitors we provide clear evidence that inhibition of HAT confers a strong preferential inhibitory effect on cell viability of undifferentiated LCSC lines when compared to their differentiated progeny. In vivo, CPTH6 is able to inhibit the growth of LCSC-derived xenografts and to reduce cancer stem cell content in treated tumors, as evidenced by marked reduction of tumor-initiating capacity in limiting dilution assays. Strikingly, the ability of CPTH6 to inhibit tubulin acetylation is also confirmed in vivo. Overall, our studies propose histone acetyltransferase inhibition as an attractive target for cancer therapy of NSCLC.
Collapse
Affiliation(s)
- Marta Di Martile
- Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Marianna Desideri
- Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Teresa De Luca
- Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Chiara Gabellini
- Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Simonetta Buglioni
- Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Adriana Eramo
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanni Sette
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Michele Milella
- Clinical and Experimental Oncology Department, Regina Elena National Cancer Institute, Rome, Italy
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, 'Sapienza' University, Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, 'Sapienza' University, Rome, Italy.,Pasteur Institute, Cenci Bolognetti Foundation, 'Sapienza' University, Rome, Italy
| | - Simone Carradori
- Department of Drug Chemistry and Technologies, 'Sapienza' University, Rome, Italy
| | - Daniela Secci
- Department of Drug Chemistry and Technologies, 'Sapienza' University, Rome, Italy
| | - Ruggero De Maria
- Scientific Director, Regina Elena National Cancer Institute, Rome, Italy
| | - Donatella Del Bufalo
- Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Daniela Trisciuoglio
- Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
7
|
Joerger M, Finn SP, Cuffe S, Byrne AT, Gray SG. The IL-17-Th1/Th17 pathway: an attractive target for lung cancer therapy? Expert Opin Ther Targets 2016; 20:1339-1356. [PMID: 27353429 DOI: 10.1080/14728222.2016.1206891] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION There is strong pharmaceutical development of agents targeting the IL-17-TH17 pathway for the treatment of psoriasis (Ps) and psoriatic arthritis (PsA). Lung cancer accounts for 28% of all cancer-related deaths worldwide, and roughly 80% of patients with newly-diagnosed non-small cell lung cancer (NSCLC) present with metastatic disease, with a poor prognosis of around 12 months. Therefore, there is a high unmet medical need for the development of new and potent systemic treatments in this deadly disease. The emergence of immunotherapies such as anti-PD-1 or anti-PDL1 as candidate therapies in non-small cell lung cancer (NSCLC) indicates that targeting critical immuno-modulatory cytokines including those within the IL-17-Th1/Th17 axis may have proven benefit in the treatment of lung cancer. Areas covered: In this review we describe the current evidence for aberrant IL-17-Th1/Th17 settings in cancer, particularly with regard to targeting this axis in NSCLC. We further discuss the current agents under pharmaceutical development which could potentially target this axis, and discuss the current limitations and areas of concern regarding the use of these in lung cancer. Expert opinion: Current evidence suggests that moving forward agents targeting the IL-17-Th1/Th17 pathway may have novel new oncoimmunology indications in the treatment paradigm for NSCLC.
Collapse
Affiliation(s)
- Markus Joerger
- a Department of Medical Oncology & Hematology , Cantonal Hospital , St. Gallen , Switzerland
| | - Stephen P Finn
- b Department of Histopathology & Morbid Anatomy , Trinity College Dublin , Dublin , Ireland
| | - Sinead Cuffe
- c HOPE Directorate , St James's Hospital , Dublin , Ireland
| | - Annette T Byrne
- d Department of Physiology and Medical Physics & Centre for Systems Medicine , Royal College of Surgeons in Ireland , Dublin , Ireland
| | - Steven G Gray
- e Thoracic Oncology Research Group , IMM, St James's Hospital , Dublin , Ireland.,f Department of Clinical Medicine , Trinity College Dublin , Dublin , Ireland
| |
Collapse
|
8
|
Parodi PW. Cooperative action of bioactive components in milk fat with PPARs may explain its anti-diabetogenic properties. Med Hypotheses 2016; 89:1-7. [DOI: 10.1016/j.mehy.2015.12.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 12/30/2015] [Indexed: 01/04/2023]
|
9
|
Colaianna M, Ilmjärv S, Peterson H, Kern I, Julien S, Baquié M, Pallocca G, Bosgra S, Sachinidis A, Hengstler JG, Leist M, Krause KH. Fingerprinting of neurotoxic compounds using a mouse embryonic stem cell dual luminescence reporter assay. Arch Toxicol 2016; 91:365-391. [PMID: 27015953 PMCID: PMC5225183 DOI: 10.1007/s00204-016-1690-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 03/10/2016] [Indexed: 02/05/2023]
Abstract
Identification of neurotoxic drugs and environmental chemicals is an important challenge. However, only few tools to address this topic are available. The aim of this study was to develop a neurotoxicity/developmental neurotoxicity (DNT) test system, using the pluripotent mouse embryonic stem cell line CGR8 (ESCs). The test system uses ESCs at two differentiation stages: undifferentiated ESCs and ESC-derived neurons. Under each condition, concentration–response curves were obtained for three parameters: activity of the tubulin alpha 1 promoter (typically activated in early neurons), activity of the elongation factor 1 alpha promoter (active in all cells), and total DNA content (proportional to the number of surviving cells). We tested 37 compounds from the ESNATS test battery, which includes polypeptide hormones, environmental pollutants (including methylmercury), and clinically used drugs (including valproic acid and tyrosine kinase inhibitors). Different classes of compounds showed distinct concentration–response profiles. Plotting of the lowest observed adverse effect concentrations (LOAEL) of the neuronal promoter activity against the general promoter activity or against cytotoxicity, allowed the differentiation between neurotoxic/DNT substances and non-neurotoxic controls. Reporter activity responses in neurons were more susceptible to neurotoxic compounds than the reporter activities in ESCs from which they were derived. To relate the effective/toxic concentrations found in our study to relevant in vivo concentrations, we used a reverse pharmacokinetic modeling approach for three exemplary compounds (teriflunomide, geldanamycin, abiraterone). The dual luminescence reporter assay described in this study allows high-throughput, and should be particularly useful for the prioritization of the neurotoxic potential of a large number of compounds.
Collapse
Affiliation(s)
- Marilena Colaianna
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland
| | - Sten Ilmjärv
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland
| | | | - Ilse Kern
- Department of Pediatrics, Geneva University Hospital, Geneva, Switzerland.,Department of Genetic and Laboratory Medicine, Geneva University Hospital, Centre Medical Universitaire, Rue Michel-Servet, 1211, Geneva 4, Switzerland
| | - Stephanie Julien
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland
| | | | - Giorgia Pallocca
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Constance, Germany
| | - Sieto Bosgra
- TNO, Zeist, The Netherlands.,BioMarin Pharmaceutical Inc., Leiden, The Netherlands
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, Dortmund, Germany
| | - Marcel Leist
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Constance, Germany
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland. .,Department of Genetic and Laboratory Medicine, Geneva University Hospital, Centre Medical Universitaire, Rue Michel-Servet, 1211, Geneva 4, Switzerland.
| |
Collapse
|
10
|
Khan S, Jena G. The role of butyrate, a histone deacetylase inhibitor in diabetes mellitus: experimental evidence for therapeutic intervention. Epigenomics 2015; 7:669-80. [DOI: 10.2217/epi.15.20] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The contribution of epigenetic mechanisms in diabetes mellitus (DM), β-cell reprogramming and its complications is an emerging concept. Recent evidence suggests that there is a link between DM and histone deacetylases (HDACs), because HDAC inhibitors promote β-cell differentiation, proliferation, function and improve insulin resistance. Moreover, gut microbes and diet-derived products can alter the host epigenome. Furthermore, butyrate and butyrate-producing microbes are decreased in DM. Butyrate is a short-chain fatty acid produced from the fermentation of dietary fibers by microbiota and has been proven as an HDAC inhibitor. The present review provides a pragmatic interpretation of chromatin-dependent and independent complex signaling/mechanisms of butyrate for the treatment of Type 1 and Type 2 DM, with an emphasis on the promising strategies for its drugability and therapeutic implication.
Collapse
Affiliation(s)
- Sabbir Khan
- Facility for Risk Assessment & Intervention Studies, Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Sector-67, S.A.S. Nagar, Punjab 60 062, India
| | - Gopabandhu Jena
- Facility for Risk Assessment & Intervention Studies, Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Sector-67, S.A.S. Nagar, Punjab 60 062, India
| |
Collapse
|
11
|
Lee P, Murphy B, Miller R, Menon V, Banik NL, Giglio P, Lindhorst SM, Varma AK, Vandergrift WA, Patel SJ, Das A. Mechanisms and clinical significance of histone deacetylase inhibitors: epigenetic glioblastoma therapy. Anticancer Res 2015; 35:615-625. [PMID: 25667438 PMCID: PMC6052863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Glioblastoma is the most common and deadliest of malignant primary brain tumors (Grade IV astrocytoma) in adults. Current standard treatments have been improving but patient prognosis still remains unacceptably devastating. Glioblastoma recurrence is linked to epigenetic mechanisms and cellular pathways. Thus, greater knowledge of the cellular, genetic and epigenetic origin of glioblastoma is the key for advancing glioblastoma treatment. One rapidly growing field of treatment, epigenetic modifiers; histone deacetylase inhibitors (HDACis), has now shown much promise for improving patient outcomes through regulation of the acetylation states of histone proteins (a form of epigenetic modulation) and other non-histone protein targets. HDAC inhibitors have been shown, in a pre-clinical setting, to be effective anticancer agents via multiple mechanisms, by up-regulating expression of tumor suppressor genes, inhibiting oncogenes, inhibiting tumor angiogenesis and up-regulating the immune system. There are many HDAC inhibitors that are currently in pre-clinical and clinical stages of investigation for various types of cancers. This review will explain the theory of epigenetic cancer therapy, identify HDAC inhibitors that are being investigated for glioblastoma therapy, explain the mechanisms of therapeutic effects as demonstrated by pre-clinical and clinical studies and describe the current status of development of these drugs as they pertain to glioblastoma therapy.
Collapse
Affiliation(s)
- Philip Lee
- Department of Neurology and Neurosurgery & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC, U.S.A
| | - Ben Murphy
- Department of Neurology and Neurosurgery & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC, U.S.A
| | - Rickey Miller
- Department of Neurology and Neurosurgery & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC, U.S.A
| | - Vivek Menon
- Department of Neurology and Neurosurgery & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC, U.S.A
| | - Naren L Banik
- Department of Neurology and Neurosurgery & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC, U.S.A. Ralph H. Johnson VA Medical Center, Charleston, SC, U.S.A
| | - Pierre Giglio
- Department of Neurology and Neurosurgery & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC, U.S.A. Department of Neurological Surgery Ohio State University Wexner Medical College, Columbus, OH, U.S.A
| | - Scott M Lindhorst
- Department of Neurology and Neurosurgery & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC, U.S.A
| | - Abhay K Varma
- Department of Neurology and Neurosurgery & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC, U.S.A
| | - William A Vandergrift
- Department of Neurology and Neurosurgery & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC, U.S.A
| | - Sunil J Patel
- Department of Neurology and Neurosurgery & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC, U.S.A
| | - Arabinda Das
- Department of Neurology and Neurosurgery & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC, U.S.A.
| |
Collapse
|
12
|
Ding N, Ping L, Feng L, Zheng X, Song Y, Zhu J. Histone deacetylase 6 activity is critical for the metastasis of Burkitt's lymphoma cells. Cancer Cell Int 2014; 14:139. [PMID: 25546298 PMCID: PMC4276069 DOI: 10.1186/s12935-014-0139-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/22/2014] [Indexed: 11/11/2022] Open
Abstract
Background Burkitt’s lymphoma is an aggressive malignancy with high risk of metastasis to extranodal sites, such as bone marrow and central nervous system. The prognosis of metastatic Burkitt’s lymphoma is poor. Here we sought to identify a role of histone deacetylase 6 (HDAC6) in the metastasis of Burkitt’s lymphoma cells. Methods Burkitt’s lymphoma cells were pharmacologically treated with niltubacin, tubacin or sodium butyrate (NaB) or transfected with siRNAs to knock down the expression of HDAC6. Cell migration and invasion ability were measured by transwell assay, and cell cycle progression was analyzed by flow cytometry. Cell adhesion and proliferation was determined by CellTiter-Glo luminescent cell viability assay kit. Cell morphological alteration and microtubule stability were analyzed by immunofluorescence staining. Effect of niltubacin, tubacin and NaB on acetylated tubulin and siRNA efficacy were measured by western blotting. Results Suppression of histone deacetylase 6 activity significantly compromised the migration and invasion of Burkitt’s lymphoma cells, without affecting cell proliferation and cell cycle progression. Mechanistic study revealed that HDAC6 modulated chemokine induced cell shape elongation and cell adhesion probably through its action on microtubule dynamics. Conclusions We identified a critical role of HDAC6 in the metastasis of Burkitt’s lymphoma cells, suggesting that pharmacological inhibition of HDAC6 could be a promising strategy for the management of metastatic Burkitt’s lymphoma.
Collapse
Affiliation(s)
- Ning Ding
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, No.52 Fucheng Road, Haidian District, Beijing, 100142 China
| | - Lingyan Ping
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, No.52 Fucheng Road, Haidian District, Beijing, 100142 China
| | - Lixia Feng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, No.52 Fucheng Road, Haidian District, Beijing, 100142 China
| | - Xiaohui Zheng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, No.52 Fucheng Road, Haidian District, Beijing, 100142 China
| | - Yuqin Song
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, No.52 Fucheng Road, Haidian District, Beijing, 100142 China
| | - Jun Zhu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, No.52 Fucheng Road, Haidian District, Beijing, 100142 China
| |
Collapse
|
13
|
Khan S, Jena G. Sodium butyrate, a HDAC inhibitor ameliorates eNOS, iNOS and TGF-β1-induced fibrogenesis, apoptosis and DNA damage in the kidney of juvenile diabetic rats. Food Chem Toxicol 2014; 73:127-39. [PMID: 25158305 DOI: 10.1016/j.fct.2014.08.010] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 08/10/2014] [Accepted: 08/18/2014] [Indexed: 12/18/2022]
Abstract
Recent reports highlighted the role of histone deacetylases (HDACs) in the pathogenesis of diabetic nephropathy (DN), but the exact molecular mechanisms by which HDAC inhibitors ameliorate DN still remain unclear. The present study was aimed to investigate the renoprotective effects of sodium butyrate (NaB) in diabetes-induced renal damages, apoptosis and fibrosis in juvenile rats. Diabetes was induced by single injection of STZ (60mg/kg), whereas NaB (500mg/kg/day) was administrated for 21days by i.p. route in a pre- and post-treatment schedule. End-points of evaluation included biochemical estimation, histology, protein expression as well as apoptosis and DNA damage examinations. Post-treatment with NaB significantly decreased plasma glucose, creatinine, urea, histological alterations including the fibrosis and collagen deposition as well as decreased the HDACs activity, expression of eNOS, iNOS, α-SMA, collagen I, fibronectin, TGFβ-1, NFκB, apoptosis and DNA damage in the diabetic kidney. These results showed that NaB treatment improved the renal function and ameliorated the histological alterations, fibrosis, apoptosis and DNA damage in the kidney of juvenile rats.
Collapse
Affiliation(s)
- Sabbir Khan
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India.
| | - Gopabandhu Jena
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India.
| |
Collapse
|
14
|
Warnault V, Ron D. Chromatin remodeling: a new landscape to treat harmful alcohol-use disorders. Future Med Chem 2013; 5:2011-3. [PMID: 24215342 PMCID: PMC4072217 DOI: 10.4155/fmc.13.152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Vincent Warnault
- The Gallo Research Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Box 0663, San Francisco, CA 94143–0663, USA
| | - Dorit Ron
- The Gallo Research Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Box 0663, San Francisco, CA 94143–0663, USA
| |
Collapse
|
15
|
Staszkiewicz J, Power RA, Harkins LL, Barnes CW, Strickler KL, Rim JS, Bondioli KR, Eilersten KJ. Silencing histone deacetylase-specific isoforms enhances expression of pluripotency genes in bovine fibroblasts. Cell Reprogram 2013; 15:397-404. [PMID: 24020699 DOI: 10.1089/cell.2013.0026] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Histone deacetylases (HDACs) catalyze deacetylation of histones that results in altered transcriptional activity. Inhibitors of HDACs have been shown to induce transcriptional changes that contribute positively to reprogramming somatic cells either by nuclear transfer or inducing a pluripotent state. However, the exact molecular mechanisms whereby HDAC inhibitors function and the specificity of the HDAC isoforms in cell reprogramming are not yet fully understood. Herein, we report the ability of individual isoform-specific HDACs to modulate endogenous expression of pluripotency-associated genes in bovine somatic cells. This in vitro study showed that a transient selective depletion of HDACs resulted in elevated mRNA levels of Oct-4, Sox2, and Nanog. In particular, we found that inhibition of specific HDAC isoforms using small interfering (si) RNA significantly increased expression of Nanog, a key factor required for totipotency induced by somatic cell nuclear transfer and for maintaining pluripotency in embryonic and induced pluripotent stem cells. Our study suggests that this gene might be the most susceptible to HDAC activity inhibition. Moreover, a regulatory role of the class III HDAC, SIRT3, on an Oct4-Sox2-Nanog transcriptional network was revealed. We observed the upregulation of pluripotency-related genes by depletion of SIRT3. SIRT3 is localized to mitochondria and is associated with energy metabolism processes, suggesting metabolic changes may be linked to reprogramming in bovine fibroblasts. In conclusion, we show that targeting selective HDACs can potentially be useful to enhance reprogramming and that sirtuins may play a pivotal role in somatic cell reprogramming by upregulating an Oct4-Sox2-Nanog transcriptional network. Dedifferentiating donor somatic cells by upregulating developmentally important genes through specific knockdown of epigenetic targets, in particular HDACs, may provide a path to improving livestock cloning and the in vitro production of pluripotent cells.
Collapse
|
16
|
Harrison IF, Dexter DT. Epigenetic targeting of histone deacetylase: therapeutic potential in Parkinson's disease? Pharmacol Ther 2013; 140:34-52. [PMID: 23711791 DOI: 10.1016/j.pharmthera.2013.05.010] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 05/09/2013] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is the most common movement disorder affecting more than 4million people worldwide. The primary motor symptoms of the disease are due to degeneration of dopaminergic nigrostriatal neurons. Dopamine replacement therapies have therefore revolutionised disease management by partially controlling these symptoms. However these drugs can produce debilitating side effects when used long term and do not protect degenerating neurons against death. Recent evidence has highlighted a pathological imbalance in PD between the acetylation and deacetylation of the histone proteins around which deoxyribonucleic acid (DNA) is coiled, in favour of excessive histone deacetylation. This mechanism of adding/removing acetyl groups to histone lysine residues is one of many epigenetic regulatory processes which control the expression of genes, many of which will be essential for neuronal survival. Hence, such epigenetic modifications may have a pathogenic role in PD. It has therefore been hypothesised that if this pathological imbalance can be corrected with the use of histone deacetylase inhibiting agents then neurodegeneration observed in PD can be ameliorated. This article will review the current literature with regard to epigenetic changes in PD and the use of histone deacetylase inhibitors (HDACIs) in PD: examining the evidence of the neuroprotective effects of numerous HDACIs in cellular and animal models of Parkinsonian cell death. Ultimately answering the question: does epigenetic targeting of histone deacetylases hold therapeutic potential in PD?
Collapse
Affiliation(s)
- Ian F Harrison
- Parkinson's Disease Research Group, Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK.
| | | |
Collapse
|
17
|
Sun X, Zhang B, Hong X, Zhang X, Kong X. Histone deacetylase inhibitor, sodium butyrate, attenuates gentamicin-induced nephrotoxicity by increasing prohibitin protein expression in rats. Eur J Pharmacol 2013; 707:147-54. [PMID: 23528351 DOI: 10.1016/j.ejphar.2013.03.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 03/10/2013] [Accepted: 03/13/2013] [Indexed: 01/18/2023]
Abstract
The major purpose in our study was to investigate the effects of sodium butyrate (NaBu) on nephrotoxicity induced by gentamicin in rats and determine further whether the protective effect is mediated by modulation of prohibitin protein expression. Gentamicin was injected intraperitoneally (100 mg/kg body weight) once daily for 8 days to induce nephrotoxicity. The effect of acute and chronic treatment of sodium butyrate on nephrotoxicity induced by gentamicin was assessed. Various doses of sodium butyrate (50, 100, 200 mg/kg, i.p.) was administered 30 min prior to the daily gentamicin injection. Histological analysis was used to evaluate the lesions in kidney after gentamicin administration. Expression of prohibitin was evaluated with immunohistochemical and western blot analysis. The present study demonstrated that gentamicin treatment for 8 consecutive days significantly increased in the levels of blood urea nitrogen, creatinine, kidney injury molecule (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL) which indicated nephrotoxicity induced by gentamicin. In addition, chronic treatment with NaBu significantly attenuated gentamicin-induced nephrotoxicity by increasing activities of superoxide dismutase, catalase and reduced glutathione. Immunohistochemical studies in gentamicin-induced rats also demonstrated an increase in the levels of inducible prohibitin after treatment with sodium butyrate. Our results indicated that sodium butyrate, a histone deacetylase inhibitor, decreased gentamicin-induced nephrotoxicity by enhancing renal antioxidant enzymes activity and the expression of prohibitin protein.
Collapse
Affiliation(s)
- Xuefeng Sun
- Department of Cardiovascular Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | | | | | | | | |
Collapse
|
18
|
Tao YF, Pang L, Du XJ, Sun LC, Hu SY, Lu J, Cao L, Zhao WL, Feng X, Wang J, Wu D, Wang N, Ni J, Pan J. Differential mRNA expression levels of human histone-modifying enzymes in normal karyotype B cell pediatric acute lymphoblastic leukemia. Int J Mol Sci 2013; 14:3376-94. [PMID: 23389039 PMCID: PMC3588049 DOI: 10.3390/ijms14023376] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 01/29/2013] [Accepted: 01/30/2013] [Indexed: 11/16/2022] Open
Abstract
Histone modification enzymes regulate gene expression by altering the accessibility of promoters to transcription factors. We sought to determine whether the genes encoding histone modification enzymes are dysregulated in pediatric acute lymphoblastic leukemia (ALL). A real-time PCR array was designed, tested and used to profile the expression of 85 genes encoding histone modification enzymes in bone marrow mononuclear cells from 30 pediatric ALL patients and 20 normal controls. The expression profile of histone-modifying genes was significantly different between normal karyotype B cell pediatric ALL and normal controls. Eleven genes were upregulated in pediatric ALL, including the histone deacetylases HDAC2 and PAK1, and seven genes were downregulated, including PRMT2 and the putative tumor suppressor EP300. Future studies will seek to determine whether these genes serve as biomarkers of pediatric ALL. Ingenuity Pathway Analysis revealed that Gene Expression and Organ Morphology was the highest rated network, with 13 focus molecules (significance score = 35). Ingenuity Pathway Analysis also indicated that curcumin and miR-34 are upstream regulators of histone-modifying enzymes; future studies will seek to validate these results and examine the role of curcumin and miR-34 in leukemia. This study provides new clues into the molecular mechanisms of pediatric ALL.
Collapse
Affiliation(s)
- Yan-Fang Tao
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou 215003, Jiangsu, China; E-Mails: (Y.-F.T.); (L.P.); (S.-Y.H.); (J.L.); (L.C.); (W.-L.Z.); (X.F.); (J.W.); (D.W.); (N.W.)
| | - Li Pang
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou 215003, Jiangsu, China; E-Mails: (Y.-F.T.); (L.P.); (S.-Y.H.); (J.L.); (L.C.); (W.-L.Z.); (X.F.); (J.W.); (D.W.); (N.W.)
| | - Xiao-Juan Du
- Department of Gastroenterology, the 5th Hospital of Chinese PLA, Yinchuan 750004, Ningxia, China; E-Mail:
| | - Li-Chao Sun
- Department of Cell and Molecular Biology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing100021, China; E-Mail:
| | - Shao-Yan Hu
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou 215003, Jiangsu, China; E-Mails: (Y.-F.T.); (L.P.); (S.-Y.H.); (J.L.); (L.C.); (W.-L.Z.); (X.F.); (J.W.); (D.W.); (N.W.)
| | - Jun Lu
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou 215003, Jiangsu, China; E-Mails: (Y.-F.T.); (L.P.); (S.-Y.H.); (J.L.); (L.C.); (W.-L.Z.); (X.F.); (J.W.); (D.W.); (N.W.)
| | - Lan Cao
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou 215003, Jiangsu, China; E-Mails: (Y.-F.T.); (L.P.); (S.-Y.H.); (J.L.); (L.C.); (W.-L.Z.); (X.F.); (J.W.); (D.W.); (N.W.)
| | - Wen-Li Zhao
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou 215003, Jiangsu, China; E-Mails: (Y.-F.T.); (L.P.); (S.-Y.H.); (J.L.); (L.C.); (W.-L.Z.); (X.F.); (J.W.); (D.W.); (N.W.)
| | - Xing Feng
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou 215003, Jiangsu, China; E-Mails: (Y.-F.T.); (L.P.); (S.-Y.H.); (J.L.); (L.C.); (W.-L.Z.); (X.F.); (J.W.); (D.W.); (N.W.)
| | - Jian Wang
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou 215003, Jiangsu, China; E-Mails: (Y.-F.T.); (L.P.); (S.-Y.H.); (J.L.); (L.C.); (W.-L.Z.); (X.F.); (J.W.); (D.W.); (N.W.)
| | - Dong Wu
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou 215003, Jiangsu, China; E-Mails: (Y.-F.T.); (L.P.); (S.-Y.H.); (J.L.); (L.C.); (W.-L.Z.); (X.F.); (J.W.); (D.W.); (N.W.)
| | - Na Wang
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou 215003, Jiangsu, China; E-Mails: (Y.-F.T.); (L.P.); (S.-Y.H.); (J.L.); (L.C.); (W.-L.Z.); (X.F.); (J.W.); (D.W.); (N.W.)
| | - Jian Ni
- Translational Research Center, The Second Clinical School, Nanjing Medical University, Nanjing 210011, Jiangsu, China; E-Mail:
| | - Jian Pan
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou 215003, Jiangsu, China; E-Mails: (Y.-F.T.); (L.P.); (S.-Y.H.); (J.L.); (L.C.); (W.-L.Z.); (X.F.); (J.W.); (D.W.); (N.W.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel./Fax: +86-512-6778-8216
| |
Collapse
|
19
|
Abstract
Harmful excessive use of alcohol has a severe impact on society and it remains one of the major causes of morbidity and mortality in the population. However, mechanisms that underlie excessive alcohol consumption are still poorly understood, and thus available medications for alcohol use disorders are limited. Here, we report that changing the level of chromatin condensation by affecting DNA methylation or histone acetylation limits excessive alcohol drinking and seeking behaviors in rodents. Specifically, we show that decreasing DNA methylation by inhibiting the activity of DNA methyltransferase (DNMT) with systemic administration of the FDA-approved drug, 5-azacitidine (5-AzaC) prevents excessive alcohol use in mice. Similarly, we find that increasing histone acetylation via systemic treatment with several histone deacetylase (HDAC) inhibitors reduces mice binge-like alcohol drinking. We further report that systemic administration of the FDA-approved HDAC inhibitor, SAHA, inhibits the motivation of rats to seek alcohol. Importantly, the actions of both DNMT and HDAC inhibitors are specific for alcohol, as no changes in saccharin or sucrose intake were observed. In line with these behavioral findings, we demonstrate that excessive alcohol drinking increases DNMT1 levels and reduces histone H4 acetylation in the nucleus accumbens (NAc) of rodents. Together, our findings illustrate that DNA methylation and histone acetylation control the level of excessive alcohol drinking and seeking behaviors in preclinical rodent models. Our study therefore highlights the possibility that DNMT and HDAC inhibitors can be used to treat harmful alcohol abuse.
Collapse
|
20
|
Gray SG, Baird AM, O'Kelly F, Nikolaidis G, Almgren M, Meunier A, Dockry E, Hollywood D, Ekström TJ, Perry AS, O'Byrne KJ. Gemcitabine reactivates epigenetically silenced genes and functions as a DNA methyltransferase inhibitor. Int J Mol Med 2012; 30:1505-11. [PMID: 23007409 DOI: 10.3892/ijmm.2012.1138] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 08/06/2012] [Indexed: 11/06/2022] Open
Abstract
Gemcitabine is indicated in combination with cisplatin as first-line therapy for solid tumours including non-small cell lung cancer (NSCLC), bladder cancer and mesothelioma. Gemcitabine is an analogue of pyrimidine cytosine and functions as an anti-metabolite. Structurally, however, gemcitabine has similarities to 5-aza-2-deoxycytidine (decitabine/Dacogen®), a DNA methyltransferase inhibitor (DNMTi). NSCLC, mesothelioma and prostate cancer cell lines were treated with decitabine and gemcitabine. Reactivation of epigenetically silenced genes was examined by RT-PCR/qPCR. DNA methyltransferase activity in nuclear extracts and recombinant proteins was measured using a DNA methyl-transferase assay, and alterations in DNA methylation status were examined using methylation-specific PCR (MS-PCR) and pyrosequencing. We observe a reactivation of several epigenetically silenced genes including GSTP1, IGFBP3 and RASSF1A. Gemcitabine functionally inhibited DNA methyltransferase activity in both nuclear extracts and recombinant proteins. Gemcitabine dramatically destabilised DNMT1 protein. However, DNA CpG methylation was for the most part unaffected by gemcitabine. In conclusion, gemcitabine both inhibits and destabilises DNA methyltransferases and reactivates epigenetically silenced genes having activity equivalent to decitabine at concentrations significantly lower than those achieved in the treatment of patients with solid tumours. This property may contribute to the anticancer activity of gemcitabine.
Collapse
Affiliation(s)
- Steven G Gray
- Thoracic Oncology Research Group, Institute of Molecular Medicine, Trinity College Dublin, Dublin 2, Republic of Ireland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Cortés-Sempere M, de Miguel MP, Pernía O, Rodriguez C, de Castro Carpeño J, Nistal M, Conde E, López-Ríos F, Belda-Iniesta C, Perona R, Ibanez de Caceres I. IGFBP-3 methylation-derived deficiency mediates the resistance to cisplatin through the activation of the IGFIR/Akt pathway in non-small cell lung cancer. Oncogene 2012; 32:1274-83. [PMID: 22543588 DOI: 10.1038/onc.2012.146] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although many cancers initially respond to cisplatin (CDDP)-based chemotherapy, resistance frequently develops. Insulin-like growth factor-binding protein-3 (IGFBP-3) silencing by promoter methylation is involved in the CDDP-acquired resistance process in non-small cell lung cancer (NSCLC) patients. Our purpose is to design a translational-based profile to predict resistance in NSCLC by studying the role of IGFBP-3 in the phosphatidyl inositol 3-kinase (PI3K) signaling pathway. We have first examined the relationship between IGFBP-3 expression regulated by promoter methylation and activation of the epidermal growth factor receptor (EGFR), insulin-like growth factor-I receptor (IGFIR) and PI3K/AKT pathways in 10 human cancer cell lines and 25 NSCLC patients with known IGFBP-3 methylation status and response to CDDP. Then, to provide a helpful tool that enables clinicians to identify patients with a potential response to CDDP, we have calculated the association between our diagnostic test and the true outcome of analyzed samples in terms of cisplatin IC50; the inhibitory concentration that kills 50% of the cell population. Our results suggest that loss of IGFBP-3 expression by promoter methylation in tumor cells treated with CDDP may activate the PI3K/AKT pathway through the specific derepression of IGFIR signaling, inducing resistance to CDDP. This study also provides a predictive test for clinical practice with an accuracy and precision of 0.84 and 0.9, respectively, (P=0.0062). We present a biomarker test that could provide clinicians with a robust tool with which to decide on the use of CDDP, improving patient clinical outcomes.
Collapse
Affiliation(s)
- M Cortés-Sempere
- Instituto de Investigaciones Biomedicas CSIC/UAM, CIBER de Enfermedades Raras CIBERER, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Shenderov BA. Gut indigenous microbiota and epigenetics. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2012; 23:17195. [PMID: 23990811 PMCID: PMC3744659 DOI: 10.3402/mehd.v23i0.17195] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 02/29/2012] [Indexed: 12/13/2022]
Abstract
This review introduces and discusses data regarding fundamental and applied investigations in mammalian epigenomics and gut microbiota received over the last 10 years. Analysis of these data enabled us first to come to the conclusion that the multiple low-molecular-weight substances of indigenous gut microbiota origin should be considered one of the main endogenous factors actively participating in epigenomic mechanisms that are responsible for the mammalian genome reprograming and post-translated modifications. Gut microecological imbalance caused by various biogenic and abiogenic agents and factors can produce different epigenetic abnormalities and the onset and progression of metabolic diseases associated. The authors substantiate the necessity to create an international project 'Human Gut Microbiota and Epigenomics' that facilitates interdisciplinary collaborations among scientists and clinicians engaged in host microbial ecology, nutrition, metagenomics, epigenomics, and metabolomics investigations as well as in disease prevention and treatment. Some priority scientific and applied directions in the current omic technologies coupled with gnotobiological approaches are suggested that can open a new era in characterizing the role of the symbiotic microbiota small metabolic and signal molecules in the host epigenomics. Although the discussed subject is only at an early stage its validation can open novel approaches in drug discovery studies.
Collapse
Affiliation(s)
- Boris Arkadievich Shenderov
- Laboratory of Biology of bifidobacteria, Head of Research Group Probiotics and Functional Foods, Gabrichevsky Research Institute of Epidemiology and Microbiology, Moscow, Russia
| |
Collapse
|
23
|
Abstract
The development of IBS symptoms – altered bowel function and abdominal cramping in a subset of adult subjects exposed to severe enteric infections opened up an unprecedented opportunity to understand the etiology of this poorly understood disorder. Perhaps, for the reasons that these symptoms follow a severe enteric infection, and mucosal biopsy tissues are readily available, the focus of most studies thus far has been to show that mild/low-grade mucosal inflammation persisting after the initial infection has subsided causes the IBS symptoms. Parallel studies in non-infectious IBS patients, who did not have prior enteritis, showed similar mild mucosal inflammation. Together, these studies examined the mucosal infiltration of specific immune cells, increase of select inflammatory mediators, mast cell and enterochromaffin cell hyperplasia, and epithelial permeability. In spite of the fact that the data on these topics were not consistent among different studies and clinical trials with prednisone, fluoxetine, and ketotifen failed to provide relief of IBS symptoms, the predominant conclusions were that mild mucosal inflammation is the cause of IBS symptoms. However, the circular smooth muscle cells, and myenteric neurons are the primary regulators of gut motility function, while primary afferent neurons and CNS play essential roles in induction of visceral hypersensitivity – no explanation was provided as to how mild mucosal inflammation causes dysfunction in cells far removed. Accumulating evidence shows that mild mucosal inflammation in IBS patients is in physiological range. It has little deleterious effects on cells within its own environment and therefore it is unlikely to affect cells in the muscularis externa. This review discusses the disconnect between the focus on mild/low-grade mucosal inflammation and the potential mechanisms and molecular dysfunctions in smooth muscle cells, myenteric neurons, and primary afferent neurons that may underlie IBS symptoms.
Collapse
Affiliation(s)
- Sushil K Sarna
- Enteric Neuromuscular Disorders and Visceral Pain Center, Division of Gastroenterology, Department of Internal Medicine, The University of Texas Medical Branch at Galveston Galveston, TX, USA
| |
Collapse
|
24
|
Abstract
The development of IBS symptoms - altered bowel function and abdominal cramping in a subset of adult subjects exposed to severe enteric infections opened up an unprecedented opportunity to understand the etiology of this poorly understood disorder. Perhaps, for the reasons that these symptoms follow a severe enteric infection, and mucosal biopsy tissues are readily available, the focus of most studies thus far has been to show that mild/low-grade mucosal inflammation persisting after the initial infection has subsided causes the IBS symptoms. Parallel studies in non-infectious IBS patients, who did not have prior enteritis, showed similar mild mucosal inflammation. Together, these studies examined the mucosal infiltration of specific immune cells, increase of select inflammatory mediators, mast cell and enterochromaffin cell hyperplasia, and epithelial permeability. In spite of the fact that the data on these topics were not consistent among different studies and clinical trials with prednisone, fluoxetine, and ketotifen failed to provide relief of IBS symptoms, the predominant conclusions were that mild mucosal inflammation is the cause of IBS symptoms. However, the circular smooth muscle cells, and myenteric neurons are the primary regulators of gut motility function, while primary afferent neurons and CNS play essential roles in induction of visceral hypersensitivity - no explanation was provided as to how mild mucosal inflammation causes dysfunction in cells far removed. Accumulating evidence shows that mild mucosal inflammation in IBS patients is in physiological range. It has little deleterious effects on cells within its own environment and therefore it is unlikely to affect cells in the muscularis externa. This review discusses the disconnect between the focus on mild/low-grade mucosal inflammation and the potential mechanisms and molecular dysfunctions in smooth muscle cells, myenteric neurons, and primary afferent neurons that may underlie IBS symptoms.
Collapse
Affiliation(s)
- Sushil K Sarna
- Enteric Neuromuscular Disorders and Visceral Pain Center, Division of Gastroenterology, Department of Internal Medicine, The University of Texas Medical Branch at Galveston Galveston, TX, USA
| |
Collapse
|
25
|
Abstract
Neurological disease, and in particular neurodegenerative diseases, cause significant burdens on both patient and healthcare costs. Despite extensive research, treatment options for patients with these conditions remain limited, and generally, only provide modest symptomatic relief. Aberrant epigenetic post-translational modifications of proteins are emerging as important elements in the pathogenesis of neurological disease. Using Alzheimer’s disease and Huntington’s disease as examples in the following article, some of latest data linking both the histone code and the various proteins that regulate this code to the pathogenesis of neurological disease are discussed. The current evidence suggesting that pharmacologically targeting one such family, the histone deacetylases, may be of potential benefit in the treatment of such diseases is also discussed. Finally, some of the potential mechanisms to specifically target these proteins within the neurological setting are discussed.
Collapse
Affiliation(s)
- Steven G Gray
- Translational Cancer Research Group, Department of Clinical Medicine, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James’s Hospital, James’s Street, Dublin 8, Ireland
| |
Collapse
|
26
|
Cyr AR, Domann FE. The redox basis of epigenetic modifications: from mechanisms to functional consequences. Antioxid Redox Signal 2011; 15:551-89. [PMID: 20919933 PMCID: PMC3118659 DOI: 10.1089/ars.2010.3492] [Citation(s) in RCA: 194] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epigenetic modifications represent mechanisms by which cells may effectively translate multiple signaling inputs into phenotypic outputs. Recent research is revealing that redox metabolism is an increasingly important determinant of epigenetic control that may have significant ramifications in both human health and disease. Numerous characterized epigenetic marks, including histone methylation, acetylation, and ADP-ribosylation, as well as DNA methylation, have direct linkages to central metabolism through critical redox intermediates such as NAD(+), S-adenosyl methionine, and 2-oxoglutarate. Fluctuations in these intermediates caused by both normal and pathologic stimuli may thus have direct effects on epigenetic signaling that lead to measurable changes in gene expression. In this comprehensive review, we present surveys of both metabolism-sensitive epigenetic enzymes and the metabolic processes that may play a role in their regulation. To close, we provide a series of clinically relevant illustrations of the communication between metabolism and epigenetics in the pathogenesis of cardiovascular disease, Alzheimer disease, cancer, and environmental toxicity. We anticipate that the regulatory mechanisms described herein will play an increasingly large role in our understanding of human health and disease as epigenetics research progresses.
Collapse
Affiliation(s)
- Anthony R Cyr
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242-1181, USA
| | | |
Collapse
|
27
|
Frau M, Ladu S, Calvisi DF, Simile MM, Bonelli P, Daino L, Tomasi ML, Seddaiu MA, Feo F, Pascale RM. Mybl2 expression is under genetic control and contributes to determine a hepatocellular carcinoma susceptible phenotype. J Hepatol 2011; 55:111-9. [PMID: 21419759 DOI: 10.1016/j.jhep.2010.10.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 10/02/2010] [Accepted: 10/07/2010] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS MYBL2 is implicated in human malignancies and over expressed in hepatocellular carcinoma (HCC). We investigated Mybl2 role in the acquisition of susceptibility to HCC and tumor progression. METHODS MYBL2 mRNA and protein levels were evaluated by quantitative RT-PCR and immunoblotting, respectively. MYBL2 expression in HCC cell lines was controlled through MYBL2 cDNA or anti-MYBL2 siRNA transfection. Gene expression profile of cells transfected with MYBL2 was analyzed by microarray. RESULTS Low induction of Mybl2 and its target Clusterin mRNAs, in low-grade dysplastic nodules (DN), progressively increased in fast growing high-grade DN and HCC of F344 rats, susceptible to hepatocarcinogenesis, whereas no/lower increases occurred in slow growing lesions of resistant BN rats. Highest Mybl2 protein activation, prevalently nuclear, occurred in F344 than BN lesions. Highest Mybl2, Clusterin, Cdc2, and Cyclin B1 expression occurred in fast progressing DN and HCC of E2f1 transgenics, compared to c-Myc transgenics, and anti-Mybl2 siRNA had highest anti-proliferative and apoptogenic effects in cell lines from HCC of E2f1 transgenics. MYBL2 transfected HepG2 and Huh7 cells exhibited increased cell proliferation and G1-S and G2-M cell cycle phases. The opposite occurred when MYBL2 was silenced by specific siRNA. MYBL2 transfection in Huh7 cells led to upregulation of genes involved in signal transduction, cell proliferation, cell motility, and downregulation of oncosuppressor and apoptogenic genes. CONCLUSIONS mybl2 expression and activation are under genetic control. Mybl2 upregulation induces fast growth and progression of premalignant and malignant liver, through cell cycle deregulation and activation of genes and pathways related to tumor progression.
Collapse
Affiliation(s)
- Maddalena Frau
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
O'Byrne KJ, Baird AM, Kilmartin L, Leonard J, Sacevich C, Gray SG. Epigenetic regulation of glucose transporters in non-small cell lung cancer. Cancers (Basel) 2011; 3:1550-65. [PMID: 24212773 PMCID: PMC3757377 DOI: 10.3390/cancers3021550] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 02/09/2011] [Accepted: 02/09/2011] [Indexed: 11/18/2022] Open
Abstract
Due to their inherently hypoxic environment, cancer cells often resort to glycolysis, or the anaerobic breakdown of glucose to form ATP to provide for their energy needs, known as the Warburg effect. At the same time, overexpression of the insulin receptor in non-small cell lung cancer (NSCLC) is associated with an increased risk of metastasis and decreased survival. The uptake of glucose into cells is carried out via glucose transporters or GLUTs. Of these, GLUT-4 is essential for insulin-stimulated glucose uptake. Following treatment with the epigenetic targeting agents histone deacetylase inhibitors (HDACi), GLUT-3 and GLUT-4 expression were found to be induced in NSCLC cell lines, with minimal responses in transformed normal human bronchial epithelial cells (HBECs). Similar results for GLUT-4 were observed in cells derived from liver, muscle, kidney and pre-adipocytes. Bioinformatic analysis of the promoter for GLUT-4 indicates that it may also be regulated by several chromatin binding factors or complexes including CTCF, SP1 and SMYD3. Chromatin immunoprecipitation studies demonstrate that the promoter for GLUT-4 is dynamically remodeled in response to HDACi. Overall, these results may have value within the clinical setting as (a) it may be possible to use this to enhance fluorodeoxyglucose (18F) positron emission tomography (FDG-PET) imaging sensitivity; (b) it may be possible to target NSCLC through the use of HDACi and insulin mediated uptake of the metabolic targeting drugs such as 2-deoxyglucose (2-DG); or (c) enhance or sensitize NSCLC to chemotherapy.
Collapse
Affiliation(s)
- Kenneth J O'Byrne
- Department of Clinical Medicine, Thoracic Oncology Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James´s Hospital, Dublin 8, Ireland.
| | | | | | | | | | | |
Collapse
|
29
|
The role of epigenetics in resistance to Cisplatin chemotherapy in lung cancer. Cancers (Basel) 2011; 3:1426-53. [PMID: 24212667 PMCID: PMC3756421 DOI: 10.3390/cancers3011426] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 03/09/2011] [Accepted: 03/10/2011] [Indexed: 12/23/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common cause of cancer related death in the world. Cisplatin and carboplatin are the most commonly used cytotoxic chemotherapeutic agents to treat the disease. These agents, usually combined with drugs such as gemcitabine or pemetrexed, induce objective tumor responses in only 20-30% of patients. Aberrant epigenetic regulation of gene expression is a frequent event in NSCLC. In this article we review the emerging evidence that epigenetics and the cellular machinery involved with this type of regulation may be key elements in the development of cisplatin resistance in NSCLC.
Collapse
|
30
|
Diao JS, Xia WS, Yi CG, Wang YM, Li B, Xia W, Liu B, Guo SZ, Sun XD. Trichostatin A inhibits collagen synthesis and induces apoptosis in keloid fibroblasts. Arch Dermatol Res 2011; 303:573-80. [PMID: 21400246 DOI: 10.1007/s00403-011-1140-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 02/25/2011] [Accepted: 02/25/2011] [Indexed: 11/25/2022]
Abstract
Keloid, a fibro-proliferative benign tumor of skin, is characterized by an enriched milieu of growth factors and an abundant accumulation of extracellular matrix (ECM). Transforming growth factor (TGF)-β1 is well known as the crucial fibrogenic cytokine promoting ECM production and tissue fibrosis in keloid forming. Epigenetic modifications have been shown to play a role in the pathogenesis of cancer as well as autoimmune and inflammatory disorders. Recent publication reports epigenetic modifications in keloid fibroblasts that include an altered pattern of DNA methylation and histone acetylation. Therefore, the field of epigenetics may provide a new therapeutic idea for keloid treatment strategies. Currently, there is some evidence from experimental studies that histone deacetylase (HDAC) inhibitor Trichostatin A (TSA) causes abrogation of TGF-β1 induced collagen synthesis in skin fibroblasts. Furthermore, TSA could suppress proliferation and induce apoptosis in a broad spectrum of tumor cells both in vitro and in vivo. These findings suggest that TSA could also cause abrogation of TGF-β1 induced collagen synthesis and induce apoptosis of proliferating keloid fibroblasts.
Collapse
Affiliation(s)
- Jian-Sheng Diao
- Institute of Plastic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Targeting Huntington's disease through histone deacetylases. Clin Epigenetics 2011; 2:257-77. [PMID: 22704341 PMCID: PMC3365382 DOI: 10.1007/s13148-011-0025-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 02/06/2011] [Indexed: 12/23/2022] Open
Abstract
Huntington’s disease (HD) is a debilitating neurodegenerative condition with significant burdens on both patient and healthcare costs. Despite extensive research, treatment options for patients with this condition remain limited. Aberrant post-translational modification (PTM) of proteins is emerging as an important element in the pathogenesis of HD. These PTMs include acetylation, phosphorylation, methylation, sumoylation and ubiquitination. Several families of proteins are involved with the regulation of these PTMs. In this review, I discuss the current evidence linking aberrant PTMs and/or aberrant regulation of the cellular machinery regulating these PTMs to HD pathogenesis. Finally, I discuss the evidence suggesting that pharmacologically targeting one of these protein families the histone deacetylases may be of potential therapeutic benefit in the treatment of HD.
Collapse
|
32
|
Abstract
IMPORTANCE OF THE FIELD Reactive oxygen species (ROS) occur as natural by-products of oxygen metabolism and have important cellular functions. Normally, the cell is able to maintain an adequate balance between the formation and removal of ROS either via anti-oxidants or through the use specific enzymatic pathways. However, if this balance is disturbed, oxidative stress may occur in the cell, a situation linked to the pathogenesis of many diseases, including cancer. AREAS COVERED IN THIS REVIEW HDACs are important regulators of many oxidative stress pathways including those involved with both sensing and coordinating the cellular response to oxidative stress. In particular aberrant regulation of these pathways by histone deacetylases may play critical roles in cancer progression. WHAT THE READER WILL GAIN In this review we discuss the notion that targeting HDACs may be a useful therapeutic avenue in the treatment of oxidative stress in cancer, using chronic obstructive pulmonary disease (COPD), NSCLC and hepatocellular carcinoma (HCC) as examples to illustrate this possibility. TAKE HOME MESSAGE Epigenetic mechanisms may be an important new therapeutic avenue for targeting oxidative stress in cancer.
Collapse
Affiliation(s)
- Matthew W Lawless
- Mater Misericordiae University Hospital, University College Dublin, Centre for Liver Disease, Dublin, Ireland
| | | | | |
Collapse
|
33
|
Abstract
Preliminary therapeutic successes have prompted a new wave of clinical trials enrolling patients with myelodysplastic syndromes (MDS), using compounds with a broad range of potential mechanisms of action. This article discusses several of the agents currently in development for MDS, reviewing clinical trial data related to five classes of novel therapeutics: clofarabine, a halogenated purine nucleoside analog; ezatiostat (TLK199), a glutathione analog that indirectly activates c-Jun kinase; tipifarnib, a farnesyltransferase inhibitor; laromustine (cloretazine), an alkylating agent with a metabolite that inhibits one mechanism of DNA damage repair; and eight drugs that inhibit histone deacetylase. Although MDS are still difficult clinical problems, and most patients with MDS still succumb to disease-related complications within 3 to 5 years of diagnosis, ongoing development of novel agents promises that there will be new treatment options for patients within the next 5 to 10 years.
Collapse
|
34
|
Ocker M. Deacetylase inhibitors - focus on non-histone targets and effects. World J Biol Chem 2010; 1:55-61. [PMID: 21540990 PMCID: PMC3083950 DOI: 10.4331/wjbc.v1.i5.55] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 04/23/2010] [Accepted: 04/30/2010] [Indexed: 02/05/2023] Open
Abstract
Inhibitors of protein deacetylases have recently been established as a novel therapeutic principle for several human diseases, including cancer. The original notion of the mechanism of action of these compounds focused on the epigenetic control of transcriptional processes, especially of tumor suppressor genes, by interfering with the acetylation status of nuclear histone proteins, hence the name histone deacetylase inhibitors was coined. Yet, this view could not explain the high specificity for tumor cells and recent evidence now suggests that non-histone proteins represent major targets for protein deacetylase inhibitors and that the post-translational modification of the acetylome is involved in various cellular processes of differentiation, survival and cell death induction.
Collapse
Affiliation(s)
- Matthias Ocker
- Matthias Ocker, Institute for Surgical Research, Philipps University Marburg, Baldingerstrasse, 35033 Marburg, Germany
| |
Collapse
|
35
|
Dietz KC, Casaccia P. HDAC inhibitors and neurodegeneration: at the edge between protection and damage. Pharmacol Res 2010; 62:11-7. [PMID: 20123018 DOI: 10.1016/j.phrs.2010.01.011] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 01/15/2010] [Indexed: 12/22/2022]
Abstract
The use of histone deacetylase inhibitors (HDACIs) as a therapeutic tool for neurodegenerative disorders has been examined with great interest in the last decade. The functional response to treatment with broad-spectrum inhibitors however, has been heterogeneous: protective in some cases and detrimental in others. In this review we discuss potential underlying causes for these apparently contradictory results. Because HDACs are part of repressive complexes, the functional outcome has been characteristically attributed to enhanced gene expression due to increased acetylation of lysine residues on nucleosomal histones. However, it is important to take into consideration that the up-regulation of diverse sets of genes (i.e. pro-apoptotic and anti-apoptotic) may orchestrate different responses in diverse cell types. An alternative possibility is that broad-spectrum pharmacological inhibition may target nuclear or cytosolic HDAC isoforms, with distinct non-histone substrates (i.e. transcription factors; cytoskeletal proteins). Thus, for any given neurological disorder, it is important to take into account the effect of HDACIs on neuronal, glial and inflammatory cells and define the relative contribution of distinct HDAC isoforms to the pathological process. This review article addresses how opposing effects on distinct cell types may profoundly influence the overall therapeutic potential of HDAC inhibitors when investigating treatments for neurodegenerative disorders.
Collapse
Affiliation(s)
- Karen C Dietz
- Department of Neuroscience and Genetics & Genomics, Mount Sinai School of Medicine, One Gustave Levy Place, Box 1065, New York, NY 10029, United States
| | | |
Collapse
|
36
|
Lawless MW, O'Byrne KJ, Gray SG. Oxidative stress induced lung cancer and COPD: opportunities for epigenetic therapy. J Cell Mol Med 2009; 13:2800-21. [PMID: 19602054 PMCID: PMC4498937 DOI: 10.1111/j.1582-4934.2009.00845.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) form as a natural by-product of the normal metabolism of oxygen and play important roles within the cell. Under normal circumstances the cell is able to maintain an adequate homeostasis between the formation of ROS and its removal through particular enzymatic pathways or via antioxidants. If however, this balance is disturbed a situation called oxidative stress occurs. Critically, oxidative stress plays important roles in the pathogenesis of many diseases, including cancer. Epigenetics is a process where gene expression is regulated by heritable mechanisms that do not cause any direct changes to the DNA sequence itself, and disruption of epigenetic mechanisms has important implications in disease. Evidence is emerging that histone deacetylases (HDACs) play decisive roles in regulating important cellular oxidative stress pathways including those involved with sensing oxidative stress and those involved with regulating the cellular response to oxidative stress. In particular aberrant regulation of these pathways by HDACs may play critical roles in cancer progression. In this review we discuss the current evidence linking epigenetics and oxidative stress and cancer, using chronic obstructive pulmonary disease and non-small cell lung cancer to illustrate the importance of epigenetics on these pathways within these disease settings.
Collapse
Affiliation(s)
- Matthew W Lawless
- Centre for Liver Disease, School of Medicine and Medical Science, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| | | | | |
Collapse
|