1
|
Ashton AW, Dhanjal HK, Rossner B, Mahmood H, Patel VI, Nadim M, Lota M, Shahid F, Li Z, Joyce D, Pajkos M, Dosztányi Z, Jiao X, Pestell RG. Acetylation of nuclear receptors in health and disease: an update. FEBS J 2024; 291:217-236. [PMID: 36471658 DOI: 10.1111/febs.16695] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/17/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
Lysine acetylation is a common reversible post-translational modification of proteins that plays a key role in regulating gene expression. Nuclear receptors (NRs) include ligand-inducible transcription factors and orphan receptors for which the ligand is undetermined, which together regulate the expression of genes involved in development, metabolism, homeostasis, reproduction and human diseases including cancer. Since the original finding that the ERα, AR and HNF4 are acetylated, we now understand that the vast majority of NRs are acetylated and that this modification has profound effects on NR function. Acetylation sites are often conserved and involve both ordered and disordered regions of NRs. The acetylated residues function as part of an intramolecular signalling platform intersecting phosphorylation, methylation and other modifications. Acetylation of NR has been shown to impact recruitment into chromatin, co-repressor and coactivator complex formation, sensitivity and specificity of regulation by ligand and ligand antagonists, DNA binding, subcellular distribution and transcriptional activity. A growing body of evidence in mice indicates a vital role for NR acetylation in metabolism. Additionally, mutations of the NR acetylation site occur in human disease. This review focuses on the role of NR acetylation in coordinating signalling in normal physiology and disease.
Collapse
Affiliation(s)
- Anthony W Ashton
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
| | | | - Benjamin Rossner
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Huma Mahmood
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Vivek I Patel
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Mohammad Nadim
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Manpreet Lota
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Farhan Shahid
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Zhiping Li
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA, USA
| | - David Joyce
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Matyas Pajkos
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Zsuzsanna Dosztányi
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Xuanmao Jiao
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA, USA
| | - Richard G Pestell
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA, USA
- The Wistar Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
2
|
Sharma G, Banerjee R, Srivastava S. Molecular Mechanisms and the Interplay of Important Chronic Obstructive Pulmonary Disease Biomarkers Reveals Novel Therapeutic Targets. ACS OMEGA 2023; 8:46376-46389. [PMID: 38107961 PMCID: PMC10719921 DOI: 10.1021/acsomega.3c07480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/02/2023] [Indexed: 12/19/2023]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a progressive, age-dependent, and unmet chronic inflammatory disease of the peripheral airways, leading to difficulty in exhalation. Several biomarkers have been tested in general towards the resolution for a long time, but no apparent success was achieved. Ongoing therapies of COPD have only symptomatic relief but no cure. Reactive oxygen species (ROS) are highly reactive species which include oxygen radicals and nonradical derivatives, and are the prominent players in COPD. They are produced as natural byproducts of cellular metabolism, but their levels can vary due to exposure to indoor air pollution, occupational pollution, and environmental pollutants such as cigarette smoke. In COPD, the lungs are continuously exposed to high levels of ROS thus leading to oxidative stress. ROS can cause damage to cells, proteins, lipids, and DNA which further contributes to the chronic inflammation in COPD and exacerbates the disease condition. Excessive ROS production can overwhelm cellular antioxidant systems and act as signaling molecules that regulate cellular processes, including antioxidant defense mechanisms involving glutathione and sirtuins which further leads to cellular apoptosis, cellular senescence, inflammation, and sarcopenia. In this review paper, we focused on COPD from different perspectives including potential markers and different cellular processes such as apoptosis, cellular senescence, inflammation, sirtuins, and sarcopenia, and tried to connect the dots between them so that novel therapeutic strategies to evaluate and target the possible underlying mechanisms in COPD could be explored.
Collapse
Affiliation(s)
- Gautam Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| | | | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| |
Collapse
|
3
|
Lunke S, Maxwell S, Khurana I, K N H, Okabe J, Al-Hasani K, El-Osta A. Epigenetic evidence of an Ac/Dc axis by VPA and SAHA. Clin Epigenetics 2021; 13:58. [PMID: 33743782 PMCID: PMC7981901 DOI: 10.1186/s13148-021-01050-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Valproic acid (VPA) is one of the most commonly used anti-epileptic drugs with pharmacological actions on GABA and blocking voltage-gated ion channels. VPA also inhibits histone deacetylase (HDAC) activity. Suberoylanilide hydroxamic acid is also a member of a larger class of compounds that inhibit HDACs. At the time of this article, there are 123 active international clinical trials for VPA (also known as valproate, convulex, divalproex, and depakote) and SAHA (vorinostat, zolinza). While it is well known that VPA and SAHA influence the accumulation of acetylated lysine residues on histones, their true epigenetic complexity remains poorly understood. RESULTS Primary human cells were exposed to VPA and SAHA to understand the extent of histone acetylation (H3K9/14ac) using chromatin immunoprecipitation followed by sequencing (ChIP-seq). Because histone acetylation is often associated with modification of lysine methylation, we also examined H3K4me3 and H3K9me3. To assess the influence of the HDAC inhibitors on gene expression, we used RNA sequencing (RNA-seq). ChIP-seq reveals a distribution of histone modifications that is robust and more broadly regulated than previously anticipated by VPA and SAHA. Histone acetylation is a characteristic of the pharmacological inhibitors that influenced gene expression. Surprisingly, we observed histone deacetylation by VPA stimulation is a predominant signature following SAHA exposure and thus defines an acetylation/deacetylation (Ac/Dc) axis. ChIP-seq reveals regionalisation of histone acetylation by VPA and broader deacetylation by SAHA. Independent experiments confirm H3K9/14 deacetylation of NFκB target genes by SAHA. CONCLUSIONS The results provide an important framework for understanding the Ac/Dc axis by highlighting a broader complexity of histone modifications by the most established and efficacious anti-epileptic medication in this class, VPA and comparison with the broad spectrum HDAC inhibitor, SAHA.
Collapse
Affiliation(s)
- Sebastian Lunke
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Scott Maxwell
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Epigenetics in Human Health and Disease Laboratory, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Ishant Khurana
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Epigenetics in Human Health and Disease Laboratory, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Harikrishnan K N
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Epigenetics in Human Health and Disease Laboratory, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jun Okabe
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Epigenetics in Human Health and Disease Laboratory, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Keith Al-Hasani
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Epigenetics in Human Health and Disease Laboratory, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Assam El-Osta
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia. .,Epigenetics in Human Health and Disease Laboratory, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia. .,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia. .,Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia. .,Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR. .,Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, 3/F Lui Che Woo Clinical Sciences Building, 30-32 Ngan Shing Street, Sha Tin, Hong Kong SAR. .,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR. .,Biomedical Laboratory Science, Department of Technology, Faculty of Health, University College Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Improved embryo development using high cysteamine concentration during IVM and sperm co-culture with COCs previous to ICSI in bovine. Theriogenology 2018; 117:26-33. [PMID: 29807255 DOI: 10.1016/j.theriogenology.2018.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 04/25/2018] [Accepted: 05/15/2018] [Indexed: 11/23/2022]
Abstract
In contrast to other species, intracytoplasmic sperm injection (ICSI) in bovine remains inefficient, resulting in low embryo developmental rates. It is unclear whether such inefficiency is due to the poor response of bovine ooplasms to the injection stimulus, or to the inability of bull sperm to induce oocyte activation. In order to facilitate these events, two strategies were assessed: the use of high concentration of cysteamine [Cys] during IVM; and the selection of sperm attached to cumulus cells after incubation with COCs for ICSI. First, COCs were IVM with increasing [Cys] and subjected to IVF. Zygotes from all groups were cultured under different O2 tensions and development to blastocyst was evaluated. In a second experiment, sperm were co-cultured for 3 h with COCs and acrosome reaction was studied. Afterwards, the best IVM and IVC conditions determined on Experiment 1 were used for ICSI assay. COCs were matured for 21 h with 1 (Cys 1) or 0.1 mM Cys (Cys 0.1 groups, standard condition). In addition, COCs were incubated for ≥3 h with 16 × 106 sperm/ml and only sperm attached to cumulus cells were selected for ICSI (ICSI + Co-cult groups). After chemical activation, embryos were cultured in SOF medium under low O2 tension. Cleavage and blastocyst rates were evaluated at days 2 and 7 of IVC, respectively. Finally, the relative expression of eight genes indicators of embryo quality was compared between ICSI and IVF control blastocysts by qPCR. Cleavage rates were higher for Cys 0.1 ICSI + Co-cult and Cys 1 ICSI + Co-cult groups (n = 117, 92% and n = 116, 79%, respectively) compared to their controls (n = 132, 60% for Cys 0.1 ICSI and n = 108, 52% for Cys 1 ICSI) (p ≤ 0.05). Interestingly, the combined treatment (Cys 1 ICSI + Co-cult) showed higher blastocyst rates than all other ICSI groups (23 vs. 11, 18 and 14% for Cys 0.1 ICSI + Co-cult, Cys 1 ICSI, and Cys 0.1 ICSI, respectively) (p ≤ 0.05). Moreover, incubation with COCs increased the rates of live acrosome reacted sperm (p ≤ 0.05). The relative abundance of mRNAs coding for INFτ, CAT, DNMT1, OCT4, and HDAC3 did not differ between treatments (p ≤ 0.05). SOD2, HADC1 and HADC2 expression was higher for Cys 0.1 ICSI than for IVF embryos (p ≤ 0.05). Group Cys 1 ICSI did not differ from IVF for those three genes, neither did Cys 1 ICSI + Co-cult, except for HDAC1 (p ≤ 0.05). In conclusion, the use of 1 mM Cys during IVM and of sperm incubated with mature COCs might be a good strategy to improve ICSI outcomes in cattle.
Collapse
|
5
|
Bronchain OJ, Chesneau A, Monsoro-Burq AH, Jolivet P, Paillard E, Scanlan TS, Demeneix BA, Sachs LM, Pollet N. Implication of thyroid hormone signaling in neural crest cells migration: Evidence from thyroid hormone receptor beta knockdown and NH3 antagonist studies. Mol Cell Endocrinol 2017; 439:233-246. [PMID: 27619407 DOI: 10.1016/j.mce.2016.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 09/08/2016] [Accepted: 09/08/2016] [Indexed: 11/18/2022]
Abstract
Thyroid hormones (TH) have been mainly associated with post-embryonic development and adult homeostasis but few studies report direct experimental evidence for TH function at very early phases of embryogenesis. We assessed the outcome of altered TH signaling on early embryogenesis using the amphibian Xenopus as a model system. Precocious exposure to the TH antagonist NH-3 or impaired thyroid receptor beta function led to severe malformations related to neurocristopathies. These include pathologies with a broad spectrum of organ dysplasias arising from defects in embryonic neural crest cell (NCC) development. We identified a specific temporal window of sensitivity that encompasses the emergence of NCCs. Although the initial steps in NCC ontogenesis appeared unaffected, their migration properties were severely compromised both in vivo and in vitro. Our data describe a role for TH signaling in NCCs migration ability and suggest severe consequences of altered TH signaling during early phases of embryonic development.
Collapse
Affiliation(s)
- Odile J Bronchain
- Paris-Saclay Institute of Neuroscience, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91405, Orsay, France.
| | - Albert Chesneau
- Paris-Saclay Institute of Neuroscience, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91405, Orsay, France
| | - Anne-Hélène Monsoro-Burq
- Univ Paris Sud, Université Paris Saclay, Centre Universitaire, F-91405, Orsay, France; Institut Curie PSL Research University, Centre Universitaire, F-91405, Orsay, France; UMR 3347 CNRS, U1021 Inserm, Université Paris Saclay, Centre Universitaire, F-91405, Orsay, France
| | - Pascale Jolivet
- CNRS, Sorbonne Universités, UPMC University Paris 06, UMR8226, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, Institut de Biologie Physico-Chimique, 75005, Paris, France; UMR 7221 CNRS, Muséum National d'histoire Naturelle, Dépt. Régulation, Développement et Diversité Moléculaire, Sorbonne Universités, 75005, Paris, France
| | - Elodie Paillard
- Watchfrog S.A., 1 Rue Pierre Fontaine, 91000, Evry, France; Institute of Systems and Synthetic Biology, CNRS, Université d'Evry Val d'Essonne, Bâtiment 3, Genopole(®) Campus 3, 1, Rue Pierre Fontaine, F-91058, Evry, France
| | - Thomas S Scanlan
- Department of Physiology & Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, L334, Portland, OR, 97239-3098, USA
| | - Barbara A Demeneix
- UMR 7221 CNRS, Muséum National d'histoire Naturelle, Dépt. Régulation, Développement et Diversité Moléculaire, Sorbonne Universités, 75005, Paris, France
| | - Laurent M Sachs
- UMR 7221 CNRS, Muséum National d'histoire Naturelle, Dépt. Régulation, Développement et Diversité Moléculaire, Sorbonne Universités, 75005, Paris, France
| | - Nicolas Pollet
- Institute of Systems and Synthetic Biology, CNRS, Université d'Evry Val d'Essonne, Bâtiment 3, Genopole(®) Campus 3, 1, Rue Pierre Fontaine, F-91058, Evry, France; Evolution, Génomes, Comportement & Ecologie, CNRS, IRD, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette, France
| |
Collapse
|
6
|
The effect of ovine oocyte vitrification on expression of subset of genes involved in epigenetic modifications during oocyte maturation and early embryo development. Theriogenology 2016; 86:2136-2146. [DOI: 10.1016/j.theriogenology.2016.07.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 11/20/2022]
|
7
|
Mondal S, Raja K, Schweizer U, Mugesh G. Chemie und Biologie der Schilddrüsenhormon-Biosynthese und -Wirkung. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201601116] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Santanu Mondal
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore Indien
| | - Karuppusamy Raja
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore Indien
| | - Ulrich Schweizer
- Rheinische Friedrich-Wilhelms-Universität Bonn; Institut für Biochemie und Molekularbiologie; Nussallee 11 53115 Bonn Deutschland
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore Indien
| |
Collapse
|
8
|
Mondal S, Raja K, Schweizer U, Mugesh G. Chemistry and Biology in the Biosynthesis and Action of Thyroid Hormones. Angew Chem Int Ed Engl 2016; 55:7606-30. [DOI: 10.1002/anie.201601116] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Santanu Mondal
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore India
| | - Karuppusamy Raja
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore India
| | - Ulrich Schweizer
- Rheinische Friedrich-Wilhelms-Universität Bonn; Institut für Biochemie und Molekularbiologie; Nussallee 11 53115 Bonn Germany
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore India
| |
Collapse
|
9
|
Skelton MM, Kampira EE, Wonkam AA, Mhandire KK, Kumwenda JJ, Duri KK, Dandara CC. Frequency variation among sub-Saharan populations in virus restriction gene, BST-2 proximal promoter polymorphisms: implications for HIV-1 prevalence differences among African countries. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2014; 18:461-71. [PMID: 24601767 DOI: 10.1089/omi.2013.0127] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The present study reports promoter variants in four sub-Saharan African populations that may affect BST-2 gene regulation. Recently, an in/del within the BST-2 promoter has been associated with HIV-1 disease progression in a Spanish cohort. Hence, we sequenced the proximal promoter region of the BST-2 gene in 581 individuals from South Africa, Zimbabwe, Malawi, and Cameroon. Seven SNPs were identified: rs28413176 (+26i6/Δ6); rs28413175 (-160i1/Δ1), -187A>G (nucleotide position -17516614); rs28413174 (-193G>A); rs73921425 (-199G>A); rs12609479 (-201C>T); and rs112492472 (-225C>T). The -199A and -225T alleles showed interesting trends across the sub-Saharan continent. Using predictive bioinformatics tools, we show that allelic variation at -199 and -201 potentially affect key transcription factor binding sites including bHLH, c-Myb, and E47. Importantly, data available from the ENCODE study gave further credence to our hypothesis of transcriptional regulation of BST-2 by a bHLH TF such as Mxi1. The possible repressive transcriptional effect of Mxi1 combined with the allelic frequency trend seen at -199 between African populations overlays well with current HIV-1 prevalence data, and may be a contributing factor to this phenomenon. The differences in HIV-1 prevalence in African countries could be, in part, due to distribution of genetic variants that affect susceptibility to HIV-1. Our findings therefore have substantive value for the design of future diagnostics for global health oriented diagnostics for HIV-1 susceptibility, and rational therapeutics on the critical path to personalized medicine in the African continent. As HIV-1 epidemiology vastly impacts human populations around the world, the population genomics strategy we have utilized herein can have value for other global regions as well.
Collapse
Affiliation(s)
- Michelle M Skelton
- 1 Division of Human Genetics, Faculty of Health Sciences, University of Cape Town , Observatory, Cape Town, South Africa
| | | | | | | | | | | | | |
Collapse
|
10
|
Sundar IK, Yao H, Rahman I. Oxidative stress and chromatin remodeling in chronic obstructive pulmonary disease and smoking-related diseases. Antioxid Redox Signal 2013; 18:1956-71. [PMID: 22978694 PMCID: PMC3624634 DOI: 10.1089/ars.2012.4863] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Chronic obstructive pulmonary disease (COPD) is predominantly a tobacco smoke-triggered disease with features of chronic low-grade systemic inflammation and aging (inflammaging) of the lung associated with steroid resistance induced by cigarette smoke (CS)-mediated oxidative stress. Oxidative stress induces various kinase signaling pathways leading to chromatin modifications (histone acetylation/deacetylation and histone methylation/demethylation) in inflammation, senescence, and steroid resistance. RECENT ADVANCES Histone mono-, di-, or tri-methylation at lysine residues result in either gene activation (H3K4, H3K36, and H3K79) or repression (H3K9, H3K27, and H3K20). Cross-talk occurs between various epigenetic marks on histones and DNA methylation. Both CS and oxidants alter histone acetylation/deacetylation and methylation/demethylation leading to enhanced proinflammatory gene expression. Chromatin modifications occur in lungs of patients with COPD. Histone deacetylase 2 (HDAC2) reduction (levels and activity) is associated with steroid resistance in response to oxidative stress. CRITICAL ISSUES Histone modifications are associated with DNA damage/repair and epigenomic instability as well as premature lung aging, which have implications in the pathogenesis of COPD. HDAC2/SIRTUIN1 (SIRT1)-dependent chromatin modifications are associated with DNA damage-induced inflammation and senescence in response to CS-mediated oxidative stress. FUTURE DIRECTIONS Understanding CS/oxidative stress-mediated chromatin modifications and the cross-talk between histone acetylation and methylation will demonstrate the involvement of epigenetic regulation of chromatin remodeling in inflammaging. This will lead to identification of novel epigenetic-based therapies against COPD and other smoking-related lung diseases. Pharmacological activation of HDAC2/SIRT1 or reversal of their oxidative post-translational modifications may offer therapies for treatment of COPD and CS-related diseases based on epigenetic histone modifications.
Collapse
Affiliation(s)
- Isaac K Sundar
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | |
Collapse
|
11
|
Gediya LK, Njar VC. Promise and challenges in drug discovery and development of hybrid anticancer drugs. Expert Opin Drug Discov 2013; 4:1099-111. [PMID: 23480431 DOI: 10.1517/17460440903341705] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Because cancer is a complex disease, it is unlikely that a single mono functional 'targeted' drug will be effective for treating this most advanced disease. Combined drugs that impact multiple targets simultaneously are better at controlling complex disease systems, are less prone to drug resistance and are the standard of care in cancer treatment. In order to improve the efficiency of using a two-drug cocktail, one approach involves the use of the so-called hybrid drugs, which comprises the incorporation of two drugs in a single molecule with the intention of exerting dual drug action. OBJECTIVE In the present article, we discuss the design, synthesis and various applications of anticancer hybrid agents and the developments in this field during the last few decades. Additionally, we describe different types of linkers and their role in contributing towards biological effects and the in vivo mechanism of drug release. We also depict some challenges from scientific and regulatory perspectives in the hybrid drug development process. CONCLUSION In the era of increasing drug resistance in cancer patients, the discovery of hybrid drugs could provide an effective strategy to create chemical entities likely to be more efficacious and less prone to resistance. However, some technical and regulatory challenges will have to be surmounted before hybrid drugs succeed in the clinical settings and justify the considerable promise of this novel concept.
Collapse
Affiliation(s)
- Lalji K Gediya
- Research Instructor, Thomas Jefferson University, Jefferson School of Pharmacy, Department of Pharmaceutical Sciences, 130 South 9th Street, Edison Building, Suite 1510 F, Philadelphia, PA 19107, USA
| | | |
Collapse
|
12
|
Reynolds N, O'Shaughnessy A, Hendrich B. Transcriptional repressors: multifaceted regulators of gene expression. Development 2013; 140:505-12. [DOI: 10.1242/dev.083105] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Through decades of research it has been established that some chromatin-modifying proteins can repress transcription, and thus are generally termed ‘repressors’. Although classic repressors undoubtedly silence transcription, genome-wide studies have shown that many repressors are associated with actively transcribed loci and that this is a widespread phenomenon. Here, we review the evidence for the presence of repressors at actively transcribed regions and assess what roles they might be playing. We propose that the modulation of expression levels by chromatin-modifying, co-repressor complexes provides transcriptional fine-tuning that drives development.
Collapse
Affiliation(s)
- Nicola Reynolds
- Wellcome Trust – Medical Research Council Stem Cell Institute, Department of Biochemistry, University of Cambridge, Cambridge CB2 1QRUK
| | - Aoife O'Shaughnessy
- Wellcome Trust – Medical Research Council Stem Cell Institute, Department of Biochemistry, University of Cambridge, Cambridge CB2 1QRUK
| | - Brian Hendrich
- Wellcome Trust – Medical Research Council Stem Cell Institute, Department of Biochemistry, University of Cambridge, Cambridge CB2 1QRUK
| |
Collapse
|
13
|
Grimaldi A, Buisine N, Miller T, Shi YB, Sachs LM. Mechanisms of thyroid hormone receptor action during development: lessons from amphibian studies. Biochim Biophys Acta Gen Subj 2012; 1830:3882-92. [PMID: 22565053 DOI: 10.1016/j.bbagen.2012.04.020] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 03/28/2012] [Accepted: 04/21/2012] [Indexed: 12/17/2022]
Abstract
BACKGROUND Thyroid hormone (TH) receptor (TR) plays critical roles in vertebrate development. However, the in vivo mechanism of TR action remains poorly explored. SCOPE OF REVIEW Frog metamorphosis is controlled by TH and mimics the postembryonic period in mammals when high levels of TH are also required. We review here some of the findings on the developmental functions of TH and TR and the associated mechanisms obtained from this model system. MAJOR CONCLUSION A dual function model for TR in Anuran development was proposed over a decade ago. That is, unliganded TR recruits corepressors to TH response genes in premetamorphic tadpoles to repress these genes and prevent premature metamorphic changes. Subsequently, when TH becomes available, liganded TR recruits coactivators to activate these same genes, leading to metamorphic changes. Over the years, molecular and genetic approaches have provided strong support for this model. Specifically, it has been shown that unliganded TR recruits histone deacetylase containing corepressor complexes during larval stages to control metamorphic timing, while liganded TR recruits multiple histone modifying and chromatin remodeling coactivator complexes during metamorphosis. These complexes can alter chromatin structure via nucleosome position alterations or eviction and histone modifications to contribute to the recruitment of transcriptional machinery and gene activation. GENERAL SIGNIFICANCE The molecular mechanisms of TR action in vivo as revealed from studies on amphibian metamorphosis are very likely applicable to mammalian development as well. These findings provide a new perspective for understanding the diverse effects of TH in normal physiology and diseases caused by TH dysfunction. This article is part of a Special Issue entitled Thyroid hormone signalling.
Collapse
Affiliation(s)
- Alexis Grimaldi
- Muséum National d'Histoire Naturelle, Dépt. Régulation Développement et Diversité Moléculaire, UMR7221 CNRS, Evolution des Régulations Endocriniennes, Section on thyroid hormone receptor function and mechanism of action, 57 rue Cuvier, 75231 Paris cedex 05, France
| | | | | | | | | |
Collapse
|
14
|
Khoo KHP, Able AJ, Chataway TK, Able JA. Preliminary characterisation of two early meiotic wheat proteins after identification through 2D gel electrophoresis proteomics. FUNCTIONAL PLANT BIOLOGY : FPB 2012; 39:222-235. [PMID: 32480776 DOI: 10.1071/fp11253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 12/24/2011] [Indexed: 06/11/2023]
Abstract
Various genetic-based approaches including mutant population screens, microarray analyses, cloning and transgenesis have broadened our knowledge of gene function during meiosis in plants. Nonetheless, these genetic tools are not without inherent limitations. One alternative approach to studying plant meiosis, especially in polyploids such as Triticum aestivum L. (bread wheat), is proteomics. However, protein-based approaches using proteomics have seldom been described, with only two attempts at studying early plant meiosis reported. Here, we report the investigation of early bread wheat meiosis using proteomics. Five differentially expressed protein spots were identified using 2D gel electrophoresis (2DGE) on protein extracts from four pooled stages of meiosis and three genotypes (Chinese Spring wild-type, ph1b and ph2a wheat mutant lines). Tandem mass spectrometry (MS/MS) identification of peptides from these protein spots led to the isolation and characterisation of the full-length clones of a wheat Speckle-type POZ protein, an SF21-like protein and HSP70, and a partial coding sequence of a hexose transporter. Significantly, the putative functions of the Speckle-type POZ protein and HSP70 were confirmed using in vitro DNA binding assays. Through the use of a 2DGE proteomics approach, we show that proteomics is a viable alternative to genetic-based approaches when studying meiosis in wheat. More significantly, we report a potential role for a Speckle-type POZ protein and a HSP70 in chromosome pairing during the early stages of meiosis in bread wheat.
Collapse
Affiliation(s)
- Kelvin H P Khoo
- School of Agriculture, Food and Wine, Waite Research Institute, The University of Adelaide, Waite Campus, PMB1, Glen Osmond, SA 5064, Australia
| | - Amanda J Able
- School of Agriculture, Food and Wine, Waite Research Institute, The University of Adelaide, Waite Campus, PMB1, Glen Osmond, SA 5064, Australia
| | - Timothy K Chataway
- Proteomics Laboratory, School of Medicine, Flinders University, Bedford Park, SA 5042, Australia
| | - Jason A Able
- School of Agriculture, Food and Wine, Waite Research Institute, The University of Adelaide, Waite Campus, PMB1, Glen Osmond, SA 5064, Australia
| |
Collapse
|
15
|
Wong CYA, Wuriyanghan H, Xie Y, Lin MF, Abel PW, Tu Y. Epigenetic regulation of phosphatidylinositol 3,4,5-triphosphate-dependent Rac exchanger 1 gene expression in prostate cancer cells. J Biol Chem 2011; 286:25813-22. [PMID: 21636851 PMCID: PMC3138273 DOI: 10.1074/jbc.m110.211292] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Revised: 05/17/2011] [Indexed: 12/21/2022] Open
Abstract
Aberrant up-regulation of P-Rex1 expression plays important roles in cancer progression and metastasis. The present study investigated the regulatory mechanism underlying P-Rex1 gene expression in prostate cancer cells. We showed that P-Rex1 expression was much higher in metastatic prostate cancer cells than in prostate epithelial cells and non-metastatic prostate cancer cells. Histone deacetylase (HDAC) inhibitors or silence of endogenous HDAC1 and HDAC2 markedly elevated P-Rex1 transcription in non-metastatic prostate cancer cells, whereas overexpression of recombinant HDAC1 in metastatic prostate cancer cells suppressed P-Rex1 expression. HDAC inhibitor trichostatin A (TSA) also significantly increased P-Rex1 promoter activity and caused acetylated histones to accumulate and associate with the P-Rex1 promoter. One Sp1 site, essential for basal promoter activity, was identified as critical for the TSA effect. TSA treatment did not alter the DNA-binding activity of Sp1 toward the P-Rex1 promoter; however, it facilitated the dissociation of the repressive HDAC1 and HDAC2 from the Sp1 binding region. Interestingly, HDAC1 association with Sp1 and with the P-Rex1 promoter were much weaker in metastatic prostate cancer PC-3 cells than in non-metastatic prostate cancer cells, and HDAC inhibitors only had very modest stimulatory effects on P-Rex1 promoter activity and P-Rex1 expression in PC-3 cells. Altogether, our studies demonstrate that HDACs could regulate P-Rex1 gene transcription by interaction with Sp1 and by region-specific changes in histone acetylation within the P-Rex1 promoter. Disassociation of HDACs from Sp1 on the P-Rex1 promoter may contribute to aberrant up-regulation of P-Rex1 in cancer.
Collapse
Affiliation(s)
- Chuu-Yun A. Wong
- From the Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska 68178 and
| | - Hada Wuriyanghan
- From the Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska 68178 and
| | - Yan Xie
- From the Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska 68178 and
| | - Ming-Fong Lin
- the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68131
| | - Peter W. Abel
- From the Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska 68178 and
| | - Yaping Tu
- From the Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska 68178 and
| |
Collapse
|
16
|
Bilesimo P, Jolivet P, Alfama G, Buisine N, Le Mevel S, Havis E, Demeneix BA, Sachs LM. Specific histone lysine 4 methylation patterns define TR-binding capacity and differentiate direct T3 responses. Mol Endocrinol 2011; 25:225-37. [PMID: 21239616 DOI: 10.1210/me.2010-0269] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The diversity of thyroid hormone T(3) effects in vivo makes their molecular analysis particularly challenging. Indeed, the current model of the action of T(3) and its receptors on transcription does not reflect this diversity. Here, T(3)-dependent amphibian metamorphosis was exploited to investigate, in an in vivo developmental context, how T(3) directly regulates gene expression. Two, direct positively regulated T(3)-response genes encoding transcription factors were analyzed: thyroid hormone receptor β (TRβ) and TH/bZIP. Reverse transcription-real-time quantitative PCR analysis on Xenopus tropicalis tadpole brain and tail fin showed differences in expression levels in premetamorphic tadpoles (lower for TH/bZIP than for TRβ) and differences in induction after T(3) treatment (lower for TRβ than for TH/bZIP). To dissect the mechanisms underlying these differences, chromatin immunoprecipitation was used. T(3) differentially induced RNA polymerase II and histone tail acetylation as a function of transcriptional level. Gene-specific patterns of TR binding were found on the different T(3) -responsive elements (higher for TRβ than for TH/bZIP), correlated with gene-specific modifications of H3K4 methylation (higher for TRβ than for TH/bZIP). Moreover, tissue-specific modifications of H3K27 were found (lower in brain than in tail fin). This first in vivo analysis of the association of histone modifications and TR binding/gene activation during vertebrate development for any nuclear receptor indicate that chromatin context of thyroid-responsive elements loci controls the capacity to bind TR through variations in histone H3K4 methylation, and that the histone code, notably H3, contributes to the fine tuning of gene expression that underlies complex physiological T(3) responses.
Collapse
Affiliation(s)
- Patrice Bilesimo
- Muséum National d’Histoire Naturelle, Départment Régulation Développement et Diversité Moléculaire, Unité Mixte de Recherche 7221 Centre National de la Recherche Scientifique, Evolution des régulations endocriniennes, 75231 Paris cedex 05, France
| | | | | | | | | | | | | | | |
Collapse
|
17
|
LEE MJ, KIM SW, LEE HG, IM GS, YANG BC, KIM NH, KIM DH. Trichostatin A Promotes the Development of Bovine Somatic Cell Nuclear Transfer Embryos. J Reprod Dev 2011; 57:34-42. [DOI: 10.1262/jrd.10-012a] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Min-Jung LEE
- National Institute of Animal Science
- Department of Animal Sciences, Chungbuk National University
| | | | | | - Gi-Sun IM
- National Institute of Animal Science
| | | | - Nam-Hyung KIM
- Department of Animal Sciences, Chungbuk National University
| | | |
Collapse
|
18
|
Sundar IK, Caito S, Yao H, Rahman I. Oxidative stress, thiol redox signaling methods in epigenetics. Methods Enzymol 2010; 474:213-44. [PMID: 20609913 DOI: 10.1016/s0076-6879(10)74013-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Epigenetics is referred to as heritable changes in gene expression but not encoded in the DNA sequence itself which occurs during posttranslational modifications in DNA and histones. These epigenetic modifications include histone acetylation, deacetylation, and methylation. Acetylation by histone acetyltransferases (HATs) of specific lysine residues on the N-terminal tail of core histones results in uncoiling of the DNA and increased accessibility to transcription factor binding. In contrast, histone deacetylation by histone deacetylases (HDACs) represses gene transcription by promoting DNA winding thereby limiting access to transcription factors. Reactive oxygen species (ROS) are involved in cellular redox alterations, such as amplification of proinflammatory and immunological responses, signaling pathways, activation of transcription factors (NF-kappaB and AP-1), chromatin remodeling (histone acetylation and deacetylation), histone/protein deacetylation by sirtuin 1 (SIRT1) and gene expression. The glutathione redox status plays an important role in protein modifications and signaling pathways, including effects on redox-sensitive transcription factors. Protein S-glutathiolation and mixed disulfide formation as candidate mechanisms for protein regulation during intracellular oxidative stress have gained a renewed impetus in view of their involvements in redox regulation of signaling proteins. A variety of methods are applied to study the epigenetic processes to elucidate the molecular mysteries underlying epigenetic inheritance. These include chromatin immunoprecipitation (ChIP), which is a powerful tool to study protein-DNA interaction and is widely used in many fields to study protein associated with chromatin, such as histone and its isoforms and transcription factors, across a defined DNA domain. Here, we describe some of the contemporary methods used to study oxidative stress and thiol redox signaling involved in epigenetic (histone acetylation, deacetylation, and methylation) and chromatin remodeling (HAT, HDAC, SIRT1) research.
Collapse
Affiliation(s)
- Isaac K Sundar
- Lung Biology and Disease Program, Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | | | | | | |
Collapse
|
19
|
Nalvarte I, Schwend T, Gustafsson JA. Proteomics analysis of the estrogen receptor alpha receptosome. Mol Cell Proteomics 2010; 9:1411-22. [PMID: 20348541 DOI: 10.1074/mcp.m900457-mcp200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The estrogen receptors (ERs) are ligand-dependent transcription factors that activate transcription by binding to estrogen response elements. Estrogen-mediated effects are tissue- and cell type-specific, determined by the cofactor recruitment to the ERs among other factors. To understand these differences in estrogen action, it is important to identify the various compositions of the ER complexes (ER receptosomes). In this report, we describe a fast and efficient method for the isolation of the ERalpha receptosome for proteomics analysis. Using immobilized estrogen response element on a Sepharose column in combination with two-dimensional electrophoresis and MALDI-TOF MS, significant amounts of proteins could be isolated and identified. Differences in ERalpha complex composition with the ER ligands 17beta-estradiol, 4-hydroxytamoxifen, and ICI-182,780 could also be observed. Thus, this approach provides an easy and relevant way of identifying ERalpha cofactor and transcription factor recruitment under different conditions.
Collapse
Affiliation(s)
- Ivan Nalvarte
- Department of Biosciences and Nutrition, Karolinska Institute, SE-14183 Huddinge, Sweden.
| | | | | |
Collapse
|
20
|
Kang MR, Kang JS, Han SB, Kim JH, Kim DM, Lee K, Lee CW, Lee KH, Lee CH, Han G, Kang JS, Kim HM, Park SK. A novel delta-lactam-based histone deacetylase inhibitor, KBH-A42, induces cell cycle arrest and apoptosis in colon cancer cells. Biochem Pharmacol 2009; 78:486-94. [PMID: 19445901 DOI: 10.1016/j.bcp.2009.05.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 05/04/2009] [Accepted: 05/05/2009] [Indexed: 12/17/2022]
Abstract
In this study, we investigated the anti-tumor activity of KBH-A42 [N-hydroxy-3-(2-oxo-1-(3-phenylpropyl)-1,2,5,6-tetrahydropyridin-3-yl)propanamide], a novel synthetic histone deacetylase (HDAC) inhibitor. KBH-A42 inhibited a variety of HDAC isoforms in enzyme assays and suppressed growth of various cancer cell lines. Among the cell lines examined, colon cancer cells, including SW620, SW480 and HCT-15, were the cell types most sensitive to KBH-A42. KBH-A42 inhibition of cancer cell growth was comparable to or stronger than that of suberoylanilide hydroxamic acid (SAHA), a well-known HDAC inhibitor approved by the FDA to treat cutaneous T cell lymphomas. In SW620 cells, KBH-A42 increased the acetylation of histones, mediated cell cycle arrest (G1 arrest at low doses and G2 arrest at high doses), and induced apoptosis. The cell cycle arrest and apoptosis induced by KBH-A42 might be mediated through up-regulation of p21(Waf1) and activation of caspases, respectively. In addition, KBH-A42 inhibited SW620 tumor growth in a human tumor xenograft model. Taken together, our results indicate that KBH-A42 exerts an anti-tumor activity in vitro and in vivo and is a promising therapeutic candidate to treat human cancers.
Collapse
Affiliation(s)
- Moo Rim Kang
- Bioevaluation Center, Korea Research Institute of Bioscience and Biotechnology, Yangcheong, Ochang, Cheongwon, Chungbuk, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Jacobs FMJ, van Erp S, van der Linden AJA, von Oerthel L, Burbach JPH, Smidt MP. Pitx3 potentiates Nurr1 in dopamine neuron terminal differentiation through release of SMRT-mediated repression. Development 2009; 136:531-40. [PMID: 19144721 DOI: 10.1242/dev.029769] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In recent years, the meso-diencephalic dopaminergic (mdDA) neurons have been extensively studied for their association with Parkinson's disease. Thus far, specification of the dopaminergic phenotype of mdDA neurons is largely attributed to the orphan nuclear receptor Nurr1. In this study, we provide evidence for extensive interplay between Nurr1 and the homeobox transcription factor Pitx3 in vivo. Both Nurr1 and Pitx3 interact with the co-repressor PSF and occupy the promoters of Nurr1 target genes in concert. Moreover, in vivo expression analysis reveals that Nurr1 alone is not sufficient to drive the dopaminergic phenotype in mdDA neurons but requires Pitx3 for full activation of target gene expression. In the absence of Pitx3, Nurr1 is kept in a repressed state through interaction with the co-repressor SMRT. Highly resembling the effect of ligand activation of nuclear receptors, recruitment of Pitx3 modulates the Nurr1 transcriptional complex by decreasing the interaction with SMRT, which acts through HDACs to keep promoters in a repressed deacetylated state. Indeed, interference with HDAC-mediated repression in Pitx3(-/-) embryos efficiently reactivates the expression of Nurr1 target genes, bypassing the necessity for Pitx3. These data position Pitx3 as an essential potentiator of Nurr1 in specifying the dopaminergic phenotype, providing novel insights into mechanisms underlying development of mdDA neurons in vivo, and the programming of stem cells as a future cell replacement therapy for Parkinson's disease.
Collapse
Affiliation(s)
- Frank M J Jacobs
- Department of Neuroscience & Pharmacology, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
22
|
Izawa M, Takekawa O, Arie T, Teraoka T, Yoshida M, Kimura M, Kamakura T. Inhibition of histone deacetylase causes reduction of appressorium formation in the rice blast fungus Magnaporthe oryzae. J GEN APPL MICROBIOL 2009; 55:489-98. [DOI: 10.2323/jgam.55.489] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
23
|
Morvan-Dubois G, Demeneix BA, Sachs LM. Xenopus laevis as a model for studying thyroid hormone signalling: from development to metamorphosis. Mol Cell Endocrinol 2008; 293:71-9. [PMID: 18657589 DOI: 10.1016/j.mce.2008.06.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Revised: 06/03/2008] [Accepted: 06/12/2008] [Indexed: 11/18/2022]
Abstract
Amphibian metamorphosis is a well-established model for dissecting the mechanisms underlying thyroid hormone (TH) action. How the pro-hormone, T(4), the active form, T(3), the deiodinases and the nuclear receptors (TRs) contribute to metamorphosis in Xenopus has been extensively investigated. Our recent work has concentrated on two key ideas in TH signalling in Xenopus: first, that there could be active roles for both liganded and unliganded receptors, and second, that ligand availability is a determining factor orchestrating these actions and is tightly controlled in target tissues. Recently, we addressed these questions at stages preceding metamorphosis, i.e. during embryogenesis, before differentiation of a functional thyroid gland. We show that repression by unliganded TR is essential to craniofacial and eye development during early development and that at these stages all three deiodinases are active. These results open new perspectives on the potential roles of TH signalling during embryogenesis.
Collapse
|
24
|
Havis E, Bilesimo P, Demeneix BA, Sachs LM. [Mechanisms underlying thyroid hormone response gene regulation during amphibian development]. ACTA ACUST UNITED AC 2008; 202:73-81. [PMID: 18547503 DOI: 10.1051/jbio:2008009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Amphibian metamorphosis is an excellent model to study the diverse effects of thyroid hormones (TH). TH modulate target gene expression via thyroid hormone receptors (TR). Generally, unliganded TR repress transcription, whereas liganded TR activate transcription. During metamorphosis, these dual effects of TR are evident. Moreover, we show that gene specific response to TH can underline the multiple effects of TH. Finally, studies of unliganded-thyroid hormone receptor function reveal a physiological role in eye development.
Collapse
Affiliation(s)
- Emmanuelle Havis
- UMR5166 CNRS, USM-501 Muséum National d'Histoire Naturelle, Département Régulation Développement et Diversité Moléculaire, CP32, 7 rue Cuvier, 75231 Paris Cedex 05, France
| | | | | | | |
Collapse
|
25
|
Andrews DM, Stokes ES, Carr GR, Matusiak ZS, Roberts CA, Waring MJ, Brady MC, Chresta CM, East SJ. Design and campaign synthesis of piperidine- and thiazole-based histone deacetylase inhibitors. Bioorg Med Chem Lett 2008; 18:2580-4. [DOI: 10.1016/j.bmcl.2008.03.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Revised: 03/13/2008] [Accepted: 03/14/2008] [Indexed: 10/22/2022]
|
26
|
Design and campaign synthesis of pyridine-based histone deacetylase inhibitors. Bioorg Med Chem Lett 2008; 18:2525-9. [DOI: 10.1016/j.bmcl.2008.03.058] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Revised: 03/18/2008] [Accepted: 03/18/2008] [Indexed: 11/17/2022]
|
27
|
Chen YD, Jiang YJ, Zhou JW, Yu QS, You QD. Identification of ligand features essential for HDACs inhibitors by pharmacophore modeling. J Mol Graph Model 2008; 26:1160-8. [DOI: 10.1016/j.jmgm.2007.10.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 10/16/2007] [Accepted: 10/16/2007] [Indexed: 10/22/2022]
|
28
|
cDNA microarray analysis of the differentially expressed genes involved in murine pre-osteoclast RAW264.7 cells proliferation stimulated by dexamethasone. Life Sci 2008; 82:135-48. [DOI: 10.1016/j.lfs.2007.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 09/22/2007] [Accepted: 10/07/2007] [Indexed: 11/21/2022]
|
29
|
Histone deacetylase inhibitors: a novel class of anti-cancer agents on its way to the market. PROGRESS IN MEDICINAL CHEMISTRY 2008; 46:205-80. [PMID: 18381127 DOI: 10.1016/s0079-6468(07)00005-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
30
|
Susick L, Veluthakal R, Suresh MV, Hadden T, Kowluru A. Regulatory roles for histone deacetylation in IL-1beta-induced nitric oxide release in pancreatic beta-cells. J Cell Mol Med 2007; 12:1571-83. [PMID: 18053091 PMCID: PMC3918073 DOI: 10.1111/j.1582-4934.2007.00171.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Histone (de)acetylases control gene transcription via modification of the chromatin structure. Herein, we investigated potential roles for histone deacetylation (or hypoacetylation) in interleukin-1β (IL-1β)-mediated inducible nitric oxide synthase (iNOS) and nitric oxide (NO) release in insulin-secreting INS 832/13 (INS) cells. Western blot analysis suggested localization of members of Class 1 and Class 2 families of histone deacetylases (HDACs) in these cells. Trichostatin A (TSA), a known inhibitor of HDACs, markedly reduced IL-1β-mediated iNOS expression and NO release from these cells in a concentration-dependent manner. TSA also promoted hyperacetylation of histone H4 under conditions in which it inhibited IL-1β-mediated effects on isolated β cells. Rottlerin, a known inhibitor of protein kinase Cδ, also increased histone H4 acetylation, and inhibited IL-1β-induced iNOS expression and NO release in these cells. It appears that the putative mechanism underlying the stimulatory effects of rottlerin on acetylation status of histone H4 are distinct from the HDAC inhibitory property of TSA, since rottlerin failed to inhibit HDAC activity in nuclear extracts isolated from INS cells. These data are suggestive of potential regulatory effects of rottlerin at the level of increasing the histone acetyltransferase activity in these cells. Together our studies present the first evidence to suggest a PKCδ-mediated signalling step, which promotes hypoacetylation of candidate histones culminating in IL-1β-induced metabolic dysfunction of the isolated β cell.
Collapse
Affiliation(s)
- L Susick
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | | | | | | | | |
Collapse
|
31
|
Depsipeptide (FK228) inhibits growth of human prostate cancer cells. Urol Oncol 2007; 26:182-9. [PMID: 18312939 DOI: 10.1016/j.urolonc.2007.01.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2006] [Revised: 01/15/2007] [Accepted: 01/18/2007] [Indexed: 11/23/2022]
Abstract
FK228 (depsipeptide) is a natural prodrug that inhibits class I histone deacetylases. We aimed to investigate the effects FK228 has on prostate cancer cells in vivo. In non-obese diabetic-severe combined immunodeficient mice implanted with human prostate cancer cells, 50 mg/kg FK228 given orally 3 times a week inhibited tumor growth and metastasis. The median time to the experimental end point (tumor volume 2 cm(3) or death) in the untreated group was 52 days, and average tumor volume was 0.8 +/- 0.18 cm(3). At the same time, 94.4% of FK228-treated mice survived and had average tumor volumes of 0.37 +/- 0.1 cm(3). All untreated animals died at 98 days, whereas, 61% of treated animal remained alive. Sizeable metastatic tumors positively stained for prostate-specific antigen (PSA), and limited air gaps were found in the lungs of untreated mice. In animals treated with FK228, lung morphology appeared normal. Primary tumors of treated animals were highly positive for PSA, and had an increased level of p21 and the proapoptotic protein Bax. Sections taken from FK228-treated animals and examined under an electron microscope showed condensed chromatin and apoptotic bodies. PSA serum levels were higher in untreated than in treated animals and correlated with tumor volume. Because prolonged oral administration of 50 mg/kg or a single oral dose of 1.2 g/kg FK228 did not cause adverse effects and inhibited proliferation of human prostate cancer cells in vivo, FK228 likely has a potential anticancer effect for prostate cancer.
Collapse
|
32
|
Elloumi HZ, Holland SM. Complex regulation of human cathelicidin gene expression: novel splice variants and 5'UTR negative regulatory element. Mol Immunol 2007; 45:204-17. [PMID: 17709140 PMCID: PMC2121615 DOI: 10.1016/j.molimm.2007.04.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 04/06/2007] [Accepted: 04/12/2007] [Indexed: 01/11/2023]
Abstract
Cationic antimicrobial peptides play important roles in host defense, linking innate and adaptive immunity. hCAP18, the only human antimicrobial cathelicidin, consists of a conserved N-terminal cathelin-like domain and a C-terminal peptide, LL-37. Expression is regulated during myeloid differentiation, and tightly controlled during infection and inflammation, suggesting active regulation. Using 5' RACE (rapid amplification of cDNA ends), multiple transcription initiation sites were identified, as well as new splice variants leading to novel augmentations of hCAP18 amino acid composition in bone marrow but not peripheral blood neutrophils. Having expressed hCAP18 promoter constructs in cell lines, we found that full-length (-1739) and truncated (-978) promoter constructs had lower luciferase activities than 5'UTR deletion constructs. Transient transfection of progressively deleted constructs in the non-permissive K562 cell line led us to identify a negative regulatory element within the 53 bp immediately upstream of the ATG of hCAP18. Additionally, transient transfection of 5' deletion constructs identified a positive regulatory element within the 101 bases 5' of promoter sequence containing two GT-boxes. Negative and positive regulatory elements within the hCAP18 gene promoter provide new insights into the possible molecular basis of myeloid gene expression.
Collapse
Affiliation(s)
- Houda Zghal Elloumi
- Immunopathogenesis Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, CRC B3-4141, MSC 1684, Bethesda, MD 20892, USA
| | | |
Collapse
|
33
|
Geng L, Cuneo KC, Fu A, Tu T, Atadja PW, Hallahan DE. Histone deacetylase (HDAC) inhibitor LBH589 increases duration of gamma-H2AX foci and confines HDAC4 to the cytoplasm in irradiated non-small cell lung cancer. Cancer Res 2007; 66:11298-304. [PMID: 17145876 DOI: 10.1158/0008-5472.can-06-0049] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Histone deacetylases (HDAC) have been identified as therapeutic targets due to their regulatory function in DNA structure and organization. LBH589 is a novel inhibitor of class I and II HDACs. We studied the effect of LBH589 and ionizing radiation (IR) on DNA repair in two human non-small cell lung cancer (NSCLC) cell lines (H23 and H460). gamma-H2AX foci present at DNA double-strand breaks (DSBs) were detected in the nuclei following 3 Gy irradiation for up to 6 hours. LBH589 administered before irradiation increased the duration of gamma-H2AX foci beyond 24 hours. Furthermore, radiation alone induced translocation of HDAC4 to the nucleus. In contrast, treatment with LBH589 followed by irradiation resulted in HDAC4 confinement to the cytoplasm, indicating that HDAC inhibition affects the nuclear localization of HDAC4. The findings that LBH589 confines HDAC4 to the cytoplasm and increases the duration of gamma-H2AX foci in irradiated cell lines suggest that HDAC4 participates in DNA damage signaling following IR. Annexin-propidium iodide flow cytometry assays, cell morphology studies, and cleaved caspase-3 Western blot analysis revealed a synergistic effect of LBH589 with IR in inducing apoptosis. Clonogenic survival showed a greater than additive effect when LBH589 was administered before irradiation compared with irradiation alone. In vivo tumor volume studies showed a growth delay of 20 days with combined treatment compared with 4 and 2 days for radiation or LBH589 alone. This study identifies HDAC4 as a biomarker of LBH589 activity and recognizes the ability of LBH589 to sensitize human NSCLC to radiation-induced DNA DSBs.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Active Transport, Cell Nucleus/radiation effects
- Animals
- Apoptosis/drug effects
- Apoptosis/radiation effects
- Carcinoma, Non-Small-Cell Lung/enzymology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line, Tumor
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cell Nucleus/radiation effects
- Cell Survival/drug effects
- Cell Survival/radiation effects
- Cytoplasm/drug effects
- Cytoplasm/enzymology
- Cytoplasm/radiation effects
- Histone Acetyltransferases/antagonists & inhibitors
- Histone Acetyltransferases/metabolism
- Histone Deacetylases/metabolism
- Histones/genetics
- Histones/metabolism
- Humans
- Hydroxamic Acids/pharmacology
- Immunoblotting
- Indoles
- Lung Neoplasms/enzymology
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Mice
- Neoplasms, Experimental/enzymology
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Panobinostat
- Repressor Proteins/metabolism
- Time Factors
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Ling Geng
- Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | | | |
Collapse
|
34
|
Huber LC, Brock M, Hemmatazad H, Giger OT, Moritz F, Trenkmann M, Distler JHW, Gay RE, Kolling C, Moch H, Michel BA, Gay S, Distler O, Jüngel A. Histone deacetylase/acetylase activity in total synovial tissue derived from rheumatoid arthritis and osteoarthritis patients. ACTA ACUST UNITED AC 2007; 56:1087-93. [PMID: 17393417 DOI: 10.1002/art.22512] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is a chronic inflammatory disorder of unknown origin. Histone deacetylase (HDA) activity is considered to play a major role in the transcriptional regulation of proinflammatory genes. We undertook this study to investigate the balance of histone acetylase and HDA activity in synovial tissue from RA patients compared with that from patients with osteoarthritis (OA) and normal controls. METHODS Activity of histone acetylases and HDAs was measured in nuclear extracts of total synovial tissue samples, which were obtained from RA and OA patients undergoing surgical joint replacement, and compared with the activity in synovial tissues from patients without arthritis. Tissue expression of HDAs 1 and 2 was quantified by Western blotting. In addition, immunohistochemistry was performed for HDA-2. RESULTS Mean+/-SEM HDA activity in synovial tissue samples derived from patients with RA was measured as 1.5+/-0.3 micromoles/microg, whereas the activity levels in OA (3.2+/-0.7 micromoles/microg) and normal (7.1+/-4.2 micromoles/microg) synovial tissue samples were significantly higher. Histone acetylase activity reached similar levels in RA and OA tissues and in normal tissues. The ratio of HDA activity to histone acetylase activity in RA synovial tissue was significantly reduced (12+/-2%) compared with that in OA synovial tissue (26+/-3%). The activity ratio in normal control samples was arbitrarily set at 100+/-40%. In addition, the tissue expression of HDA-1 and HDA-2 proteins was clearly lower in RA samples than in OA samples. CONCLUSION The balance of histone acetylase/HDA activities is strongly shifted toward histone hyperacetylation in patients with RA. These results offer novel molecular insights into the pathogenesis of the disease that might be relevant to the development of future therapeutic approaches.
Collapse
Affiliation(s)
- Lars C Huber
- Center of Experimental Rheumatology, University Hospital Zurich, and Zurich Center of Integrative Human Physiology, Zurich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Havis E, Le Mevel S, Morvan Dubois G, Shi DL, Scanlan TS, Demeneix BA, Sachs LM. Unliganded thyroid hormone receptor is essential for Xenopus laevis eye development. EMBO J 2006; 25:4943-51. [PMID: 17006540 PMCID: PMC1618110 DOI: 10.1038/sj.emboj.7601356] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2006] [Accepted: 08/23/2006] [Indexed: 01/05/2023] Open
Abstract
Thyroid hormone receptors generally activate transcription of target genes in the presence of thyroid hormone (T(3)) and repress their transcription in its absence. Here, we investigated the role of unliganded thyroid hormone receptor (TR) during vertebrate development using an amphibian model. Previous studies led to the hypothesis that before production of endogenous T(3), the presence of unliganded receptor is essential for premetamorphic tadpole growth. To test this hypothesis, we generated a Xenopus laevis TR beta mutant construct ineffective for gene repression owing to impaired corepressor NCoR recruitment. Overexpression by germinal transgenesis of the mutant receptor leads to lethality during early development with numerous defects in cranio-facial and eye development. These effects correlate with TR expression profiles at these early stages. Molecular analysis of transgenic mutants reveals perturbed expression of genes involved in eye development. Finally, treatment with iopanoic acid or NH-3, modulators of thyroid hormone action, leads to abnormal eye development. In conclusion, the data reveal a role of unliganded TR in eye development.
Collapse
Affiliation(s)
- Emmanuelle Havis
- UMR5166 CNRS, USM-501 Muséum National d'Histoire Naturelle, Dépt. Régulation, Développement et Diversité Moléculaire, Paris, France
| | - Sébastien Le Mevel
- UMR5166 CNRS, USM-501 Muséum National d'Histoire Naturelle, Dépt. Régulation, Développement et Diversité Moléculaire, Paris, France
| | - Ghislaine Morvan Dubois
- UMR5166 CNRS, USM-501 Muséum National d'Histoire Naturelle, Dépt. Régulation, Développement et Diversité Moléculaire, Paris, France
| | - De-Li Shi
- UMR7622 CNRS, Laboratoire de Biologie du Développement, Université Pierre et Marie Curie, Paris VI, Paris, France
| | - Thomas S Scanlan
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
| | - Barbara A Demeneix
- UMR5166 CNRS, USM-501 Muséum National d'Histoire Naturelle, Dépt. Régulation, Développement et Diversité Moléculaire, Paris, France
| | - Laurent M Sachs
- UMR5166 CNRS, USM-501 Muséum National d'Histoire Naturelle, Dépt. Régulation, Développement et Diversité Moléculaire, Paris, France
- UMR5166 CNRS, USM-501 Muséum National d'Histoire Naturelle, Dépt. Régulation, Développement et Diversité Moléculaire, CP 33, 7 rue Cuvier, 75231 Paris cedex 05, France. Tel.: +33 1 40 79 36 04; Fax: +33 1 40 79 36 18; E-mail:
| |
Collapse
|
36
|
Mukhopadhyay NK, Weisberg E, Gilchrist D, Bueno R, Sugarbaker DJ, Jaklitsch MT. Effectiveness of trichostatin A as a potential candidate for anticancer therapy in non-small-cell lung cancer. Ann Thorac Surg 2006; 81:1034-42. [PMID: 16488717 DOI: 10.1016/j.athoracsur.2005.06.059] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2005] [Revised: 06/15/2005] [Accepted: 06/22/2005] [Indexed: 11/26/2022]
Abstract
BACKGROUND A well-known histone deacetylase inhibitor, trichostatin A, was applied to non-small-cell lung cancer cells to determine whether inhibition of histone deacetylase leads to the production of proteins that either arrest tumor cell growth or lead to tumor cell death. METHODS Trichostatin A (0.01 to 1.0 micromol/L) was applied to one normal lung fibroblast and four non-small-cell lung cancer lines, and its effect was determined by flow cytometry, annexin-V staining, immunoprecipitation, and Western blot analysis. RESULTS Trichostatin A demonstrated tenfold greater growth inhibition in all four non-small-cell lung cancer lines compared with normal controls, with a concentration producing 50% inhibition ranging from 0.01 to 0.04 micromol/L for the tumor cell lines and 0.7 micromol/L for the normal lung fibroblast line. Trichostatin A treatment reduced the percentage of cells in S phase (10% to 23%) and increased G1 populations (10% to 40%) as determined by flow cytometry. Both annexin-V binding assay and upregulation of the protein, gelsolin (threefold to tenfold), demonstrated that the tumor cells were apoptotic, whereas normal cells were predominantly in cell cycle arrest. Trichostatin A increased histone H4 acetylation and expression of p21 twofold to 15-fold without significant effect on p16, p27, CDK2, and cyclin D1. CONCLUSIONS Collectively, these data suggest that inhibition of histone deacetylation may provide a valuable approach for lung cancer treatment. We evaluated trichostatin A as a potential candidate for anticancer therapy in non-small-cell lung cancer.
Collapse
Affiliation(s)
- Nishit K Mukhopadhyay
- Division of Thoracic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Engel D, Nudelman A, Levovich I, Gruss-Fischer T, Entin-Meer M, Phillips DR, Cutts SM, Rephaeli A. Mode of interaction between butyroyloxymethyl-diethyl phosphate (AN-7) and doxorubicin in MCF-7 and resistant MCF-7/Dx cell lines. J Cancer Res Clin Oncol 2006; 132:673-83. [PMID: 16826403 DOI: 10.1007/s00432-006-0116-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2006] [Accepted: 05/05/2006] [Indexed: 11/25/2022]
Abstract
PURPOSE To investigate the anticancer activity and mode of action of butyroyloxymethyl-diethyl phosphate (AN-7), a prodrug of butyric acid and formaldehyde, as a single agent and in combination with doxorubicin in human carcinoma MCF-7 and the multidrug resistant MCF-7 Dx cell lines. METHODS The anti-cancer activity of AN-7 as a single agent or in combination with doxorubicin was measured by the Hoechst cell viability and colony forming assays as well as by FACS analyses of cells stained with propidium iodide and annexin V-FITC. Modulations of protein expression and acetylation were measured by Western blot analyses. The number of doxorubicin-DNA adducts formed was evaluated using (14)C-labeled doxorubicin. RESULTS The AN-7 and homologous prodrugs exhibited similar growth inhibition effects against drug resistant and sensitive cells, and elicited their anticancer effect partially by inhibition of HDAC. The AN-7 transiently augmented histone acetylation and increase of p21 expression. Synergy between AN-7 and doxorubicin was demonstrated in the sensitive and the resistant cell lines by viability and colony formation assays and was further confirmed by FACS analysis showing an increase in cell mortality. The number of doxorubicin-DNA adducts in total genomic DNA isolated from cells treated with (14)C-labeled doxorubicin and AN-7 increased substantially compared to treatment with doxorubicin only. Treatment with AN-7 or doxorubicin increased p53 acetylation that was further potentiated by their combination. CONCLUSION The AN-7 combined with doxorubicin overcame drug resistance; at least in part by the intracellularly releasable formaldehyde that augmented formation of doxorubicin-DNA adducts and butyric acid that induced histone and p53 acetylation. Since the use of doxorubicin is limited by toxicity, the combination could offer an effective treatment modality with lower toxicity for breast cancer.
Collapse
Affiliation(s)
- Dikla Engel
- Chemistry Department, Bar-Ilan University, Ramat Gan, 52900, Israel
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Lee KY, Ito K, Hayashi R, Jazrawi EPI, Barnes PJ, Adcock IM. NF-kappaB and activator protein 1 response elements and the role of histone modifications in IL-1beta-induced TGF-beta1 gene transcription. THE JOURNAL OF IMMUNOLOGY 2006; 176:603-15. [PMID: 16365456 DOI: 10.4049/jimmunol.176.1.603] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Abnormal expression of TGF-beta1 is believed to play an important role in the pathogenesis of a number of chronic inflammatory and immune lung diseases, including asthma, chronic obstructive pulmonary disease, and pulmonary fibrosis. Gene activation in eukaryotes requires coordinated use of specific cell signals, chromatin modifications, and chromatin remodeling. We studied the roles of the ubiquitous inflammatory transcription factors, NF-kappaB and AP-1, in activation of the TGF-beta1 gene and histone acetylation at the TGF-beta1 promoter. IL-1beta-induced TGF-beta1 protein secretion and mRNA expression were prevented by actinomycin D and were attenuated by the inhibitor of kappaB kinase 2 inhibitor AS602868 and the JNK inhibitor SP600125, suggesting a degree of transcriptional regulation mediated by the NF-kappaB and AP-1 pathways. We demonstrated that IL-1beta activated the p65 subunit of NF-kappaB and the c-Jun subunit of AP-1. Using chromatin immunoprecipitation assays, we observed a sequential recruitment of p65 and c-Jun, accompanying ordered elevation of the levels of histone H4 and H3 acetylation and recruitment of RNA polymerase II at distinct regions in the native TGF-beta1 promoter. The specific NF-kappaB and AP-1 binding sites in the TGF-beta1 promoter were confirmed by an ELISA-based binding assay, and evidence for histone hyperacetylation in TGF-beta1 induction was supported by the observation that the histone deacetylase inhibitor trichostatin A enhanced basal and IL-1beta-induced TGF-beta1 mRNA expression. Our results suggest that IL-1beta-stimulated transcription of TGF-beta1 is temporally regulated by NF-kappaB and AP-1 and involves histone hyperacetylation at distinct promoter sites.
Collapse
Affiliation(s)
- Kang-Yun Lee
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
39
|
Ito K, Yamamura S, Essilfie-Quaye S, Cosio B, Ito M, Barnes PJ, Adcock IM. Histone deacetylase 2-mediated deacetylation of the glucocorticoid receptor enables NF-kappaB suppression. ACTA ACUST UNITED AC 2005; 203:7-13. [PMID: 16380507 PMCID: PMC2118081 DOI: 10.1084/jem.20050466] [Citation(s) in RCA: 449] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Glucocorticoids are the most effective antiinflammatory agents for the treatment of chronic inflammatory diseases even though some diseases, such as chronic obstructive pulmonary disease (COPD), are relatively glucocorticoid insensitive. However, the molecular mechanism of this glucocorticoid insensitivity remains uncertain. We show that a defect of glucocorticoid receptor (GR) deacetylation caused by impaired histone deacetylase (HDAC) 2 induces glucocorticoid insensitivity toward nuclear factor (NF)-κB–mediated gene expression. Specific knockdown of HDAC2 by RNA interference resulted in reduced sensitivity to dexamethasone suppression of interleukin 1β–induced granulocyte/macrophage colony-stimulating factor production. Loss of HDAC2 did not reduce GR nuclear translocation, GR binding to glucocorticoid response element (GRE) on DNA, or GR-induced DNA or gene induction but inhibited the association between GR and NF-κB. GR becomes acetylated after ligand binding, and HDAC2-mediated GR deacetylation enables GR binding to the NF-κB complex. Site-directed mutagenesis of K494 and K495 reduced GR acetylation, and the ability to repress NF-κB–dependent gene expression becomes insensitive to histone deacetylase inhibition. In conclusion, we show that overexpression of HDAC2 in glucocorticoid-insensitive alveolar macrophages from patients with COPD is able to restore glucocorticoid sensitivity. Thus, reduction of HDAC2 plays a critical role in glucocorticoid insensitivity in repressing NF-κB–mediated, but not GRE-mediated, gene expression.
Collapse
Affiliation(s)
- Kazuhiro Ito
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London SW3 6LY, England, UK.
| | | | | | | | | | | | | |
Collapse
|
40
|
Papeleu P, Vanhaecke T, Elaut G, Vinken M, Henkens T, Snykers S, Rogiers V. Differential effects of histone deacetylase inhibitors in tumor and normal cells-what is the toxicological relevance? Crit Rev Toxicol 2005; 35:363-78. [PMID: 15989141 DOI: 10.1080/10408440590935639] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Histone deacetylase (HDAC) inhibitors target key steps of tumor development: They inhibit proliferation, induce differentiation and/or apoptosis, and exhibit potent antimetastatic and antiangiogenic properties in transformed cells in vitro and in vivo. Preliminary studies in animal models have revealed a relatively high tumor selectivity of HDAC inhibitors, strenghtening their promising potential in cancer chemotherapy. Until now, preclinical in vitro research has almost exclusively been performed in cancer cell lines and oncogene-transformed cells. However, as cell proliferation and apoptosis are essential for normal tissue and organ homeostasis, it is important to investigate how HDAC inhibitors influence the regulation of and interplay between proliferation, differentiation, and apoptosis in primary cells as well. This review highlights the discrepancies in molecular events triggered by trichostatin A, the reference compound of hydroxamic acid-containing HDAC inhibitors, in hepatoma cells and primary hepatocytes (which are key targets for drug-induced toxicity). The implications of these differential outcomes in both cell types are discussed with respect to both toxicology and drug development. In view of the future use of HDAC inhibitors as cytostatic drugs, it is highly recommended to include both tumor cells and their healthy counterparts in preclinical developmental studies. Screening the toxicological properties of compounds early in their development process, using a battery of different cell types, will enable researchers to discard those compounds bearing undesirable adverse activity before entering into expensive clinical trials. This will not only reduce the risk for harmful exposure of patients but also save time and money.
Collapse
Affiliation(s)
- Peggy Papeleu
- Department of Toxicology, Vrije Universiteit Brussel, Brussels, Belgium.
| | | | | | | | | | | | | |
Collapse
|
41
|
Popov N, Wahlström T, Hurlin PJ, Henriksson M. Mnt transcriptional repressor is functionally regulated during cell cycle progression. Oncogene 2005; 24:8326-37. [PMID: 16103876 DOI: 10.1038/sj.onc.1208961] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The Myc/Max/Mad network of transcription factors regulates cell proliferation, differentiation, and transformation. Similar to other proteins of the network, Mnt forms heterodimers with Max and binds CACGTG E-Box elements. Transcriptional repression by Mnt is mediated through association with mSin3, and deletion of the mSin3-interacting domain (SID) converts Mnt to a transcriptional activator. Mnt is coexpressed with Myc in proliferating cells and has been suggested to be a modulator of Myc function. We report that Mnt is expressed both in growth-arrested and proliferating mouse fibroblasts and is phosphorylated when resting cells are induced to re-enter the cell cycle. Importantly, the interaction between Mnt and mSin3 is disrupted upon serum stimulation resulting in decreased Mnt-associated HDAC activity. Furthermore, we demonstrate that Mnt binds and recruits mSin3 to the Myc target gene cyclin D2 in quiescent mouse fibroblasts. Interference with Mnt expression by RNAi resulted in upregulation of cyclin D2 expression in growth-arrested fibroblasts, supporting the view that Mnt represses cyclin D2 transcription in quiescent cells. Our data suggest a model in which phosphorylation of Mnt at cell cycle entry results in disruption of Mnt-mSin3-HDAC1 interaction, which allows induction of Myc target genes by release of Mnt-mediated transcriptional repression.
Collapse
Affiliation(s)
- Nikita Popov
- Microbiology and Tumor Biology Center, Karolinska Institutet, Box 280, SE-171 77 Stockholm, Sweden
| | | | | | | |
Collapse
|
42
|
Song L, Zlobin A, Ghoshal P, Zhang Q, Houde C, Weijzen S, Jiang Q, Nacheva E, Yagan D, Davis E, Galiegue-Zouitina S, Catovsky D, Grogan T, Fisher RI, Miele L, Coignet LJ. Alteration of SMRT tumor suppressor function in transformed non-Hodgkin lymphomas. Cancer Res 2005; 65:4554-61. [PMID: 15930272 DOI: 10.1158/0008-5472.can-04-4108] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Indolent non-Hodgkin lymphomas are characterized by a prolonged phase that is typically followed by a clinical progression associated with an accelerated clinical course and short survival time. Previous studies have not identified a consistent cytogenetic or molecular abnormality associated with transformation. The development of a transformed phenotype, evolving from the original low-grade component, most likely depends on multiple genetic events, including the activation of synergistic dominant oncogenes and a loss of tumor suppressor gene functions. Complex karyotypes and relatively bad chromosome morphology are typical of transformed non-Hodgkin lymphomas, rendering complete cytogenetic analysis difficult. Here, we report the use of transformed non-Hodgkin lymphoma cell lines and primary samples to identify the involvement of the silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) gene that maps at chromosome 12q24 in transformed non-Hodgkin lymphomas. We also show that down-regulation of SMRT in the immortalized "Weinberg's model" cell lines induces transformation of the cells. Assessment of cDNA array profiles should further help us to design a working model for SMRT involvement in non-Hodgkin lymphoma transformation as a novel, nonclassical tumor suppressor.
Collapse
MESH Headings
- Apoptosis/genetics
- Cell Line, Tumor
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Chromosomes, Human, Pair 12/genetics
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- Down-Regulation
- Gene Deletion
- Gene Rearrangement
- Genes, Tumor Suppressor
- Humans
- Immunoglobulin J Recombination Signal Sequence-Binding Protein
- Lymphoma, Non-Hodgkin/genetics
- Lymphoma, Non-Hodgkin/metabolism
- Lymphoma, Non-Hodgkin/pathology
- Nuclear Proteins/genetics
- Nuclear Receptor Co-Repressor 2
- Oligonucleotide Array Sequence Analysis
- Repressor Proteins/biosynthesis
- Repressor Proteins/genetics
- Transcriptional Activation
Collapse
Affiliation(s)
- Lynda Song
- Department of Medicine, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, Illinois, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Rephaeli A, Blank-Porat D, Tarasenko N, Entin-Meer M, Levovich I, Cutts SM, Phillips DR, Malik Z, Nudelman A. In vivo andin vitro antitumor activity of butyroyloxymethyl-diethyl phosphate (AN-7), a histone deacetylase inhibitor, in human prostate cancer. Int J Cancer 2005; 116:226-35. [PMID: 15800932 DOI: 10.1002/ijc.21030] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
AN-7, a prodrug of butyric acid, induced histone hyperacetylation and differentiation and inhibited proliferation of human prostate 22Rv1 cancer cells in vitro and in vivo. In nude mice implanted with these cells, 50 mg/kg AN-7 given orally thrice a week led to inhibition of tumor growth and metastasis, tumor regression in >25% of animals and increased survival. Median time to the experimental end point (tumor volume 2 cm3 or death) in the untreated was 52 days, and average tumor volume was 0.8 +/- 0.18 cm3. At the same time, 94.4% of AN-7-treated mice survived and had average tumor volumes of 0.37 +/- 0.1 cm3. PSA expression was a useful marker for 22Rv1 lung metastasis detection. Sizeable metastases positively stained for PSA and limited air gaps were found in lungs of untreated mice. In animals treated with AN-7, lung morphology appeared normal. Primary tumors of treated animals were highly positive for PSA and had an elevated level of p21 and the proapoptotic protein Bax. Sections taken from AN-7-treated animals, examined under an electron microscope, exhibited condensed chromatin and apoptotic bodies. PSA serum levels were higher in untreated compared to treated animals and correlated with tumor volume. Since prolonged oral administration with 50 mg/kg or a single oral dose of 1.2 g/kg AN-7 did not cause adverse effects and the former exhibited significant anticancer activity, AN-7 is likely to display a high therapeutic index and may be beneficial for prostate cancer patients.
Collapse
Affiliation(s)
- Ada Rephaeli
- Faculty of Medicine, Felsenstein Center for Medical Research, Tel Aviv University Beilinson Campus, Petach Tikva, Israel.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sanderson L, Taylor GW, Aboagye EO, Alao JP, Latigo JR, Coombes RC, Vigushin DM. Plasma pharmacokinetics and metabolism of the histone deacetylase inhibitor trichostatin a after intraperitoneal administration to mice. Drug Metab Dispos 2004; 32:1132-8. [PMID: 15269190 DOI: 10.1124/dmd.104.000638] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Trichostatin A is a potent and specific histone deacetylase inhibitor with promising antitumor activity in preclinical models. Plasma pharmacokinetics of trichostatin A were studied following single-dose intraperitoneal administration of 80 mg/kg (high dose) or 0.5 mg/kg (low dose) to female BALB/c mice. Plasma trichostatin A concentrations were quantified by high performance liquid chromatography (HPLC)-UV assay (high dose) or by HPLC-multiple reaction monitoring assay (low dose). Trichostatin A was rapidly absorbed from the peritoneum and detectable in plasma within 2 min. Cmax of 40 microg/ml and 8 ng/ml occurred within 5 min, followed by rapid exponential decay in plasma trichostatin A concentration with t1/2 of 6.3 min and 9.6 min (high and low doses, respectively). Phase I metabolites at the high dose were identified by simultaneous UV and positive ion electrospray mass spectrometry. Trichostatin A underwent extensive metabolism: primary metabolic pathways were N-demethylation, reduction of the hydroxamic acid to the corresponding trichostatin A amide, and oxidative deamination to trichostatic acid. N-Monomethyl trichostatin A amide was the major plasma metabolite. No didemethylated compounds were identified. Trichostatic acid underwent further biotransformation: reduction and beta-oxidation of the carboxylic acid, with or without N-demethylation, resulted in formation of dihydro trichostatic acid and dinor dihydro trichostatic acids. HPLC fractions corresponding to trichostatin A and N-demethylated trichostatin A exhibited histone deacetylase-inhibitory activity; no other fractions were biologically active. We conclude that trichostatin A is rapidly and extensively metabolized in vivo following intraperitoneal administration to mice, and N-demethylation does not compromise histone deacetylase-inhibitory activity.
Collapse
Affiliation(s)
- L Sanderson
- Department of Cancer Medicine, 6th Floor MRC Cyclotron Building, Imperial College London (Hammersmith Hospital Campus), Du Cane Road, London W12 0NN, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
45
|
Assenat E, Gerbal-Chaloin S, Larrey D, Saric J, Fabre JM, Maurel P, Vilarem MJ, Pascussi JM. Interleukin 1beta inhibits CAR-induced expression of hepatic genes involved in drug and bilirubin clearance. Hepatology 2004; 40:951-60. [PMID: 15382119 DOI: 10.1002/hep.20387] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
During the inflammatory response, intrahepatic cholestasis and decreased drug metabolism are frequently observed. At the hepatic level, the orphan nuclear constitutive androstane receptor (CAR) (NR1I3) controls phase I (cytochrome P450 [CYP] 2B and CYP3A), phase II (UGT1A1), and transporter (SLC21A6, MRP2) genes involved in drug metabolism and bilirubin clearance in response to xenobiotics such as phenobarbital or endobiotics such as bilirubin. We investigated the negative regulation of CAR, a glucocorticoid-responsive gene, via proinflammatory cytokine interleukin 1beta (IL-1beta) and lipopolysaccharides (LPSs) in human hepatocytes. We show that IL-1beta decreases CAR expression and decreases phenobarbital- or bilirubin-mediated induction of CYP2B6, CYP2C9, CYP3A4, UGT1A1, GSTA1, GSTA2, and SLC21A6 messenger RNA. This occurs via nuclear factor kappaB (NF-kappaB) p65 activation, which interferes with the enhancer function of the distal glucocorticoid response element that we have identified recently in the CAR promoter. We demonstrate that: (1) LPSs, IL-1beta, or overexpression of p65RelA inhibit glucocorticoid receptor (GR)-mediated CAR transactivation; (2) these suppressive effects can be blocked both by pyrrolidine dithiocarbamate, an inhibitor of NF-kappaB activation, or by overexpression of SRIkBalpha, a NF-kappaB repressor; and (3) the GR agonist dexamethasone induces histone H4 acetylation at the proximal CAR promoter region, whereas LPSs and IL-1beta inhibit this acetylation as assessed via chromatin immunoprecipitation assay. In conclusion, GR/NF-kappaB interaction affects CAR gene transcription through chromatin remodeling and provide a mechanistic explanation for the long-standing observation that inflammation and sepsis inhibit drug metabolism while inducing intrahepatic cholestasis or hyperbilirubinemia.
Collapse
Affiliation(s)
- Eric Assenat
- Service de Chirurgie Digestive, Hôpital Saint Eloi, Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Chittka A, Arevalo JC, Rodriguez-Guzman M, Pérez P, Chao MV, Sendtner M. The p75NTR-interacting protein SC1 inhibits cell cycle progression by transcriptional repression of cyclin E. ACTA ACUST UNITED AC 2004; 164:985-96. [PMID: 15051733 PMCID: PMC2172053 DOI: 10.1083/jcb.200301106] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Schwann cell factor 1 (SC1), a p75 neurotrophin receptor–interacting protein, is a member of the positive regulatory/suppressor of variegation, enhancer of zeste, trithorax (PR/SET) domain-containing zinc finger protein family, and it has been shown to be regulated by serum and neurotrophins. SC1 shows a differential cytoplasmic and nuclear distribution, and its presence in the nucleus correlates strongly with the absence of bromodeoxyuridine (BrdU) in these nuclei. Here, we investigated potential transcriptional activities of SC1 and analyzed the function of its various domains. We show that SC1 acts as a transcriptional repressor when it is tethered to Gal4 DNA-binding domain. The repressive activity requires a trichostatin A–sensitive histone deacetylase (HDAC) activity, and SC1 is found in a complex with HDACs 1, 2, and 3. Transcriptional repression exerted by SC1 requires the presence of its zinc finger domains and the PR domain. Additionally, these two domains are involved in the efficient block of BrdU incorporation by SC1. The zinc finger domains are also necessary to direct SC1's nuclear localization. Lastly, SC1 represses the promoter of a promitotic gene, cyclin E, suggesting a mechanism for how growth arrest is regulated by SC1.
Collapse
Affiliation(s)
- Alexandra Chittka
- Institute for Clinical Neurobiology, University of Würzburg, Würzburg, Germany.
| | | | | | | | | | | |
Collapse
|
47
|
Naruse Y, Oh-hashi K, Iijima N, Naruse M, Yoshioka H, Tanaka M. Circadian and light-induced transcription of clock gene Per1 depends on histone acetylation and deacetylation. Mol Cell Biol 2004; 24:6278-87. [PMID: 15226430 PMCID: PMC434252 DOI: 10.1128/mcb.24.14.6278-6287.2004] [Citation(s) in RCA: 163] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Circadian clock genes are regulated through a transcriptional-translational feedback loop. Alterations of the chromatin structure by histone acetyltransferases and histone deacetylases (HDACs) are commonly implicated in the regulation of gene transcription. However, little is known about the transcriptional regulation of mammalian clock genes by chromatin modification. Here, we show that the state of acetylated histones fluctuated in parallel with the rhythm of mouse Per1 (mPer1) or mPer2 expression in fibroblast cells and liver. Mouse CRY1 (mCRY1) repressed transcription with HDACs and mSin3B, which was relieved by the HDAC inhibitor trichostatin A (TSA). In turn, TSA induced endogenous mPer1 expression as well as the acetylation of histones H3 and H4, which interacted with the mPer1 promoter region in fibroblast cells. Moreover, a light pulse stimulated rapid histone acetylation associated with the promoters of mPer1 or mPer2 in the suprachiasmatic nucleus (SCN) and the binding of phospho-CREB in the CRE of mPer1. We also showed that TSA administration into the lateral ventricle induced mPer1 and mPer2 expression in the SCN. Taken together, these data indicate that the rhythmic transcription and light induction of clock genes are regulated by histone acetylation and deacetylation.
Collapse
Affiliation(s)
- Yoshihisa Naruse
- Department of Anatomy, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-0841, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Côté S, McNamara S, Brambilla D, Bianchini A, Rizzo G, del Rincón SV, Grignani F, Nervi C, Miller WH. Expression of SMRTbeta promotes ligand-induced activation of mutated and wild-type retinoid receptors. Blood 2004; 104:4226-35. [PMID: 15319284 DOI: 10.1182/blood-2003-10-3583] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nuclear receptors are ligand-modulated transcription factors regulated by interactions with corepressors and coactivators, whose functions are not fully understood. Acute promyelocytic leukemia (APL) is characterized by a translocation, t(15;17), that produces a PML/RARalpha fusion oncoprotein, whose abnormal transcriptional function is successfully targeted by pharmacologic levels of all-trans-retinoic acid (ATRA). Mutations in the ligand-binding domain of PML/RARalpha that confer resistance to ATRA have been studied by expression in nonhematopoietic cells, such as Cos-1. Here, we show that ATRA binding and transcriptional activation by the same PML/RARalpha mutant differ markedly between nonhematopoietic and leukemic cell lines. Differential expression of the corepressor isoform silencing mediator for retinoid and thyroid receptors beta (SMRTbeta) correlates with increased ligand binding and transcription by the mutant PML/RARalpha. Transient and stable overexpression of SMRTbeta in hematopoietic cells that only express SMRTalpha increased ATRA binding, ligand-induced transcription, and ATRA-induced cell differentiation. This effect may not be limited to abnormal nuclear receptors, because overexpression of SMRTbeta increased ATRA-induced binding and transcriptional activation of wild-type receptors PML/RARalpha and RARalpha. Our results suggest a novel role for the SMRTbeta isoform whereby its cell-specific expression may influence the binding and transcriptional capacities of nuclear receptors, thus providing new evidence of distinct functions of corepressor isoforms and adding complexity to transcriptional regulation.
Collapse
MESH Headings
- Cell Line, Tumor
- Chromosomes, Human, Pair 15
- Chromosomes, Human, Pair 17
- DNA-Binding Proteins/genetics
- Drug Resistance, Neoplasm
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Jurkat Cells
- Leukemia, Promyelocytic, Acute/genetics
- Ligands
- Neoplasm Proteins/genetics
- Nuclear Receptor Co-Repressor 2
- Oncogene Proteins, Fusion/genetics
- Plasmids
- Receptors, Retinoic Acid/genetics
- Repressor Proteins/genetics
- Transcriptional Activation
- Translocation, Genetic
- Tretinoin/pharmacokinetics
- Tretinoin/toxicity
Collapse
Affiliation(s)
- Sylvie Côté
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, 3755, Chemin de la Côte Ste-Catherine, Montreal, Quebec, Canada H3T 1E2
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Liu XF, Bagchi MK. Recruitment of distinct chromatin-modifying complexes by tamoxifen-complexed estrogen receptor at natural target gene promoters in vivo. J Biol Chem 2004; 279:15050-8. [PMID: 14722073 DOI: 10.1074/jbc.m311932200] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tamoxifen, a breast cancer therapeutic, is a tissue-selective estrogen receptor modulator (SERM), which acts as an antiestrogen in the mammary tissue and displays estrogenic activity in other tissues such as bone and uterus. In order to understand the mechanisms underlying the antiestrogenic effect of this prototype SERM, we performed an analysis of the cofactors that interact with ER complexed with 4-hydroxytamoxifen (OHT) at natural target genes in a human breast tumor cell line MCF-7. Employing chromatin immunoprecipitation (ChIP), we observed that treatment with OHT rapidly induces the binding of ERalpha to the E-responsive promoter regions of pS2 and c-myc genes. Promoter-bound OHT-complexed ERa coordinately recruited the components of a multiprotein complex containing the corepressor NCoR, histone deacetylase 3 (HDAC3), and a WD40-repeat protein TBL1. Surprisingly, the OHT-complexed ERalpha also recruited a chromatin-remodeling NuRD complex in which histone deacetylase 1 (HDAC1) is associated with several polypeptides including metastasis-associated protein 1/2 (MTA1/2), and SWI2/SNF2-related ATPase Mi2. Kinetic studies revealed that following OHT addition the recruitment of these HDAC complexes to pS2 or the c-myc promoter occurs in a sequential manner; the NCoR-HDAC3 complex is recruited earlier than the NuRD complex. Serial ChIP experiments indicated that the ER-NCoR-HDAC3 and ER-NuRD complexes are distinct, and they do not occupy the target gene promoter simultaneously. We also established a close temporal link between the appearance of the HDAC complexes at the E-responsive regions of pS2 and c-myc promoters, local hypoacetylation of specific lysine residues in N-terminal tails of histones H3 and H4, and disappearance of RNA polymerase II from the target gene loci. Collectively, our studies indicated that transcriptional repression by tamoxifen-bound ER at E-regulated gene promoters involves a dynamic interplay of multiple distinct chromatin-modifying/remodeling complexes.
Collapse
Affiliation(s)
- Xue-Feng Liu
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | |
Collapse
|
50
|
Poon APW, Liang Y, Roizman B. Herpes simplex virus 1 gene expression is accelerated by inhibitors of histone deacetylases in rabbit skin cells infected with a mutant carrying a cDNA copy of the infected-cell protein no. 0. J Virol 2004; 77:12671-8. [PMID: 14610189 PMCID: PMC262575 DOI: 10.1128/jvi.77.23.12671-12678.2003] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
An earlier report showed that the expression of viral genes by a herpes simplex virus 1 mutant [HSV-1(vCPc0)] in which the wild-type, spliced gene encoding infected-cell protein no. 0 (ICP0) was replaced by a cDNA copy is dependent on both the cell type and multiplicity of infection. At low multiplicities of infection, viral gene expression in rabbit skin cells was delayed by many hours, although ultimately virus yield was comparable to that of the wild-type virus. This defect was rescued by replacement of the cDNA copy with the wild-type gene. To test the hypothesis that the delay reflected a dysfunction of ICP0 in altering the structure of host protein-viral DNA complexes, we examined the state of histone deacetylases (HDACs) (HDAC1, HDAC2, and HDAC3). We report the following. (i) HDAC1 and HDAC2, but not HDAC3, were modified in infected cells. The modification was mediated by the viral protein kinase U(S)3 and occurred between 3 and 6 h after infection with wild-type virus but was delayed in rabbit skin cells infected with HSV-1(vCPc0) mutant, concordant with a delay in the expression of viral genes. (ii) Pretreatment of rabbit skin cells with inhibitors of HDAC activity (e.g., sodium butyrate, Helminthosporium carbonum toxin, or trichostatin A) accelerated the expression of HSV-1(vCPc0) but not that of wild-type virus. We conclude the following. (i) In the interval in which HSV-1(vCPc0) DNA is silent, its DNA is in chromatin-like structures amenable to modification by inhibitors of histone deacetylases. (ii) Expression of wild-type virus genes in these cells precluded the formation of DNA-protein structures that would be affected by either the HDACs or their inhibitors. (iii) Since the defect in HSV-1(vCPc0) maps to ICP0, the results suggest that this protein initiates the process of divestiture of viral DNA from tight chromatin structures but could be replaced by other viral proteins in cells infected with a large number of virions.
Collapse
Affiliation(s)
- Alice P W Poon
- The Marjorie B. Kovler Viral Oncology Laboratories, The University of Chicago, Chicago, Illinois 60637, USA
| | | | | |
Collapse
|