51
|
Abstract
Epigenetic regulatory mechanisms play key roles in cardiac development, differentiation, homeostasis, response to stress and injury, and disease. Human heart failure (HF) epigenetic regulatory mechanisms have not been deciphered to date. This 2-part review distills the rapidly evolving research focused on human HF epigenetic regulatory mechanisms. Part I, which was published in the September/October issue, focused on epigenetic regulatory mechanisms involving RNA, specifically the role of short, intermediate, and long noncoding RNAs (lncRNAs) and endogenous competing RNA regulatory networks. Part II, now in the November/December issue, focuses on the epigenetic regulatory mechanisms involving DNA, including DNA methylation, histone modifications, and chromatin conformational changes. Part II concludes with 2 examples of well-studied integrated epigenetic regulatory mechanisms: the structural and functional roles of the Mediator complex in regulating transcription and the epigenetic networked "cross-talk" regulating atrial natriuretic peptide and brain natriuretic peptide promoter activation.
Collapse
|
52
|
Movassaghian S, Xie Y, Hildebrandt C, Rosati R, Li Y, Kim NH, Conti DS, da Rocha SRP, Yang ZQ, Merkel OM. Post-Transcriptional Regulation of the GASC1 Oncogene with Active Tumor-Targeted siRNA-Nanoparticles. Mol Pharm 2016; 13:2605-21. [PMID: 27223606 DOI: 10.1021/acs.molpharmaceut.5b00948] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Basal-like breast cancer (BLBC) accounts for the most aggressive types of breast cancer, marked by high rates of relapse and poor prognoses and with no effective clinical therapy yet. Therefore, investigation of new targets and treatment strategies is more than necessary. Here, we identified a receptor that can be targeted in BLBC for efficient and specific siRNA mediated gene knockdown of therapeutically relevant genes such as the histone demethylase GASC1, which is involved in multiple signaling pathways leading to tumorigenesis. Breast cancer and healthy breast cell lines were compared regarding transferrin receptor (TfR) expression via flow cytometry and transferrin binding assays. Nanobioconjugates made of low molecular weight polyethylenimine (LMW-PEI) and transferrin (Tf) were synthesized to contain a bioreducible disulfide bond. siRNA complexation was characterized by condensation assays and dynamic light scattering. Cytotoxicity, transfection efficiency, and the targeting specificity of the conjugates were investigated in TfR positive and negative healthy breast and breast cancer cell lines by flow cytometry, confocal microscopy, RT-PCR, and Western blot. Breast cancer cell lines revealed a significantly higher TfR expression than healthy breast cells. The conjugates efficiently condensed siRNA into particles with 45 nm size at low polymer concentrations, showed no apparent toxicity on different breast cancer cell lines, and had significantly greater transfection and gene knockdown activity on mRNA and protein levels than PEI/siRNA leading to targeted and therapeutic growth inhibition post GASC1 knockdown. The synthesized nanobioconjugates improved the efficiency of gene transfer and targeting specificity in transferrin receptor positive cells but not in cells with basal receptor expression. Therefore, these materials in combination with our newly identified siRNA sequences are promising candidates for therapeutic targeting of hard-to-treat BLBC and are currently further investigated regarding in vivo targeting efficacy and biocompatibility.
Collapse
Affiliation(s)
- Sara Movassaghian
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Oncology, Karmanos Cancer Institute, Wayne State University , Detroit, Michigan 48201, United States
| | - Yuran Xie
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States
| | - Claudia Hildebrandt
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Pharmaceutics and Biopharmaceutics, Kiel University , 24118 Kiel, Germany
| | - Rayna Rosati
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Oncology, Karmanos Cancer Institute, Wayne State University , Detroit, Michigan 48201, United States
| | - Ying Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States
| | - Na Hyung Kim
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States
| | - Denise S Conti
- Department of Chemical Engineering and Materials Science, College of Engineering, Wayne State University , Detroit, Michigan 48202, United States
| | - Sandro R P da Rocha
- Department of Pharmaceutics, College of Pharmacy, Virginia Commonwealth University , Richmond, Virginia 23298, United States.,Department of Chemical and Life Science Engineering, Virginia Commonwealth University , Richmond, Virginia 23284, United States
| | - Zeng-Quan Yang
- Department of Oncology, Karmanos Cancer Institute, Wayne State University , Detroit, Michigan 48201, United States
| | - Olivia M Merkel
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Oncology, Karmanos Cancer Institute, Wayne State University , Detroit, Michigan 48201, United States.,Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München , 80539 Munich, Germany
| |
Collapse
|
53
|
Takashina T, Kinoshita I, Kikuchi J, Shimizu Y, Sakakibara-Konishi J, Oizumi S, Nishimura M, Dosaka-Akita H. Combined inhibition of EZH2 and histone deacetylases as a potential epigenetic therapy for non-small-cell lung cancer cells. Cancer Sci 2016; 107:955-62. [PMID: 27116120 PMCID: PMC4946723 DOI: 10.1111/cas.12957] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 04/02/2016] [Accepted: 04/18/2016] [Indexed: 12/31/2022] Open
Abstract
Recent discoveries have revealed that human cancer involves aberrant epigenetic alterations. We and others have previously shown that the histone methyltransferase EZH2, the catalytic subunit of polycomb repressive complex 2 (PRC2), is frequently overexpressed in non‐small‐cell lung cancer (NSCLC) and that an EZH2 inhibitor, 3‐deazaneplanocin A, inhibits the proliferation of NSCLC cells. Transcriptional silencing by EZH2 was recently shown to be required for the activity of histone deacetylases (HDACs) that interact with another PRC2 protein, EED. To develop a more effective epigenetic therapy for NSCLC, we determined the effects of co‐treatment with 3‐deazaneplanocin A and the HDAC inhibitor vorinostat (SAHA) in NSCLC cells. The co‐treatment synergistically suppressed the proliferation of all tested NSCLC cell lines, regardless of their epidermal growth factor receptor (EGFR) status. The synergistic effect was associated with slightly decreased histone H3 lysine 27 trimethylation, modestly increased histone acetylation, and the depletion of EZH2 and other PRC2 proteins. The co‐treatment resulted in an accumulation of p27Kip1, decrease in cyclin A, and increased apoptotic fraction in an additive/synergistic manner. Interestingly, the co‐treatment strongly suppressed EGFR signaling, not only in EGFR‐wild‐type NSCLC cells, but also in EGFR‐mutant cells, mainly through dephosphorylation of EGFR. Furthermore, the co‐treatment suppressed the in vivo tumor growth of EGFR‐mutant, EGFR–tyrosine kinase‐resistant H1975 cells more effectively than did each agent alone, without visible toxicity. These results suggest that the combined pharmacological targeting of EZH2 and HDACs may provide more effective epigenetic therapeutics for NSCLC.
Collapse
Affiliation(s)
- Taichi Takashina
- First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Japan
| | - Ichiro Kinoshita
- Department of Medical Oncology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Junko Kikuchi
- First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Japan
| | - Yasushi Shimizu
- Department of Medical Oncology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | - Satoshi Oizumi
- First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Japan
| | - Masaharu Nishimura
- First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Japan
| | - Hirotoshi Dosaka-Akita
- Department of Medical Oncology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
54
|
Yong KW, Wan Safwani WKZ, Xu F, Wan Abas WAB, Choi JR, Pingguan-Murphy B. Cryopreservation of Human Mesenchymal Stem Cells for Clinical Applications: Current Methods and Challenges. Biopreserv Biobank 2016; 13:231-9. [PMID: 26280501 DOI: 10.1089/bio.2014.0104] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Mesenchymal stem cells (MSCs) hold many advantages over embryonic stem cells (ESCs) and other somatic cells in clinical applications. MSCs are multipotent cells with strong immunosuppressive properties. They can be harvested from various locations in the human body (e.g., bone marrow and adipose tissues). Cryopreservation represents an efficient method for the preservation and pooling of MSCs, to obtain the cell counts required for clinical applications, such as cell-based therapies and regenerative medicine. Upon cryopreservation, it is important to preserve MSCs functional properties including immunomodulatory properties and multilineage differentiation ability. Further, a biosafety evaluation of cryopreserved MSCs is essential prior to their clinical applications. However, the existing cryopreservation methods for MSCs are associated with notable limitations, leading to a need for new or improved methods to be established for a more efficient application of cryopreserved MSCs in stem cell-based therapies. We review the important parameters for cryopreservation of MSCs and the existing cryopreservation methods for MSCs. Further, we also discuss the challenges to be addressed in order to preserve MSCs effectively for clinical applications.
Collapse
Affiliation(s)
- Kar Wey Yong
- 1 Department of Biomedical Engineering, Faculty of Engineering, University of Malaya , Kuala Lumpur, Malaysia .,2 Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an, P.R. China
| | | | - Feng Xu
- 2 Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an, P.R. China .,3 The Key Library of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University , Xi'an, P.R. China
| | - Wan Abu Bakar Wan Abas
- 1 Department of Biomedical Engineering, Faculty of Engineering, University of Malaya , Kuala Lumpur, Malaysia
| | - Jane Ru Choi
- 1 Department of Biomedical Engineering, Faculty of Engineering, University of Malaya , Kuala Lumpur, Malaysia .,2 Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an, P.R. China
| | - Belinda Pingguan-Murphy
- 1 Department of Biomedical Engineering, Faculty of Engineering, University of Malaya , Kuala Lumpur, Malaysia
| |
Collapse
|
55
|
Abstract
The formation of spontaneous epialleles is poorly understood. A new study describes how the formation of epihybrids can lead to the appearance of novel epialleles.
Collapse
Affiliation(s)
- William T Jordan
- Department of Genetics, University of Georgia, 120 East Green Street, Athens, GA, 30602, USA
| | - Robert J Schmitz
- Department of Genetics, University of Georgia, 120 East Green Street, Athens, GA, 30602, USA.
| |
Collapse
|
56
|
Kidd CDA, Thompson PJ, Barrett L, Baltic S. Histone Modifications and Asthma. The Interface of the Epigenetic and Genetic Landscapes. Am J Respir Cell Mol Biol 2016; 54:3-12. [PMID: 26397168 DOI: 10.1165/rcmb.2015-0050tr] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Complex lung diseases, such as asthma, are influenced by both genetic predisposition and environmental stimuli. The epigenetic landscape of such diseases is attracting increasing interest and research. Epigenetics broadly covers the transient and the inheritable changes to gene expression that are not directly due to changes in nucleotide sequences. Epigenetic mechanisms could have significant impact on asthma-related allergic, immune, and regulatory pathways, as well as on the generation of biomarkers and the heritable transmission of asthma phenotypes. Recent technological advances have allowed mapping of the epigenome and analysis of genome-wide epigenetic contributors to disease. As a result, ground-breaking observations regarding histone post-translational modifications in a number of immunological diseases have emerged. In this review, we look beyond the biological information coded by DNA and review the epigenetic modifications made to histones, with evidence suggesting a role for their modification in asthma.
Collapse
Affiliation(s)
- Courtney D A Kidd
- 1 Institute for Respiratory Health, Perth, Western Australia, Australia.,2 Centre for Respiratory Health, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia; and
| | - Philip J Thompson
- 1 Institute for Respiratory Health, Perth, Western Australia, Australia.,2 Centre for Respiratory Health, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia; and.,3 Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Western Australia, Perth, Western Australia, Australia
| | - Lucy Barrett
- 1 Institute for Respiratory Health, Perth, Western Australia, Australia.,2 Centre for Respiratory Health, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia; and
| | - Svetlana Baltic
- 1 Institute for Respiratory Health, Perth, Western Australia, Australia.,2 Centre for Respiratory Health, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia; and
| |
Collapse
|
57
|
Lulla RR, Saratsis AM, Hashizume R. Mutations in chromatin machinery and pediatric high-grade glioma. SCIENCE ADVANCES 2016; 2:e1501354. [PMID: 27034984 PMCID: PMC4803494 DOI: 10.1126/sciadv.1501354] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/04/2016] [Indexed: 05/24/2023]
Abstract
Pediatric central nervous system tumors are the most common solid tumor of childhood. Of these, approximately one-third are gliomas that exhibit diverse biological behaviors in the unique context of the developing nervous system. Although low-grade gliomas predominate and have favorable outcomes, up to 20% of pediatric gliomas are high-grade. These tumors are a major contributor to cancer-related morbidity and mortality in infants, children, and adolescents, with long-term survival rates of only 10 to 15%. The recent discovery of somatic oncogenic mutations affecting chromatin regulation in pediatric high-grade glioma has markedly improved our understanding of disease pathogenesis, and these findings have stimulated the development of novel therapeutic approaches targeting epigenetic regulators for disease treatment. We review the current perspective on pediatric high-grade glioma genetics and epigenetics, and discuss the emerging and experimental therapeutics targeting the unique molecular abnormalities present in these deadly childhood brain tumors.
Collapse
Affiliation(s)
- Rishi R. Lulla
- Department of Pediatrics—Hematology, Oncology, Neuro-Oncology and Stem Cell Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Amanda Muhs Saratsis
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rintaro Hashizume
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
58
|
High-throughput small molecule screen identifies inhibitors of aberrant chromatin accessibility. Proc Natl Acad Sci U S A 2016; 113:3018-23. [PMID: 26929321 DOI: 10.1073/pnas.1521827113] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mutations in chromatin-modifying proteins and transcription factors are commonly associated with a wide variety of cancers. Through gain- or loss-of-function, these mutations may result in characteristic alterations of accessible chromatin, indicative of shifts in the landscape of regulatory elements genome-wide. The identification of compounds that reverse a specific chromatin signature could lead to chemical probes or potential therapies. To explore whether chromatin accessibility could serve as a platform for small molecule screening, we adapted formaldehyde-assisted isolation of regulatory elements (FAIRE), a chemical method to enrich for nucleosome-depleted genomic regions, as a high-throughput, automated assay. After demonstrating the validity and robustness of this approach, we applied this method to screen an epigenetically targeted small molecule library by evaluating regions of aberrant nucleosome depletion mediated by EWSR1-FLI1, the chimeric transcription factor critical for the bone and soft tissue tumor Ewing sarcoma. As a class, histone deacetylase inhibitors were greatly overrepresented among active compounds. These compounds resulted in diminished accessibility at targeted sites by disrupting transcription of EWSR1-FLI1. Capitalizing on precise differences in chromatin accessibility for drug discovery efforts offers significant advantages because it does not depend on the a priori selection of a single molecular target and may detect novel biologically relevant pathways.
Collapse
|
59
|
Lahiri S, Sun N, Buck A, Imhof A, Walch A. MALDI imaging mass spectrometry as a novel tool for detecting histone modifications in clinical tissue samples. Expert Rev Proteomics 2016; 13:275-84. [DOI: 10.1586/14789450.2016.1146598] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
60
|
Lahiri S, Sun N, Solis-Mezarino V, Fedisch A, Ninkovic J, Feuchtinger A, Götz M, Walch A, Imhof A. In situ detection of histone variants and modifications in mouse brain using imaging mass spectrometry. Proteomics 2016; 16:437-47. [PMID: 26593131 DOI: 10.1002/pmic.201500345] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/14/2015] [Accepted: 10/16/2015] [Indexed: 01/25/2023]
Abstract
Histone posttranslational modifications and histone variants control the epigenetic regulation of gene expression and affect a wide variety of biological processes. A complex pattern of such modifications and variants defines the identity of cells within complex organ systems and can therefore be used to characterize cells at a molecular level. However, their detection and identification in situ has been limited so far due to lack of specificity, selectivity, and availability of antihistone antibodies. Here, we describe a novel MALDI imaging MS based workflow, which enables us to detect and characterize histones by their intact mass and their correlation with cytological properties of the tissue using novel statistical and image analysis tools. The workflow allows us to characterize the in situ distribution of the major histone variants and their modification in the mouse brain. This new analysis tool is particularly useful for the investigation of expression patterns of the linker histone H1 variants for which suitable antibodies are so far not available.
Collapse
Affiliation(s)
- Shibojyoti Lahiri
- Protein Analysis Unit (ZfP), Biomedical Center (BMC), Ludwig Maximilians University Munich, Planegg-Martinsried, Germany
| | - Na Sun
- Research Unit Analytical Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Andreas Fedisch
- Protein Analysis Unit (ZfP), Biomedical Center (BMC), Ludwig Maximilians University Munich, Planegg-Martinsried, Germany
| | - Jovica Ninkovic
- Institute of Stem Cell Research, GSF-National Research Center for Environment and Health, Neuherberg, Germany.,Munich Cluster for Systems Neurology (SyNergy), Ludwig Maximilians University of Munich, Munich, Germany.,Institute of Physiological Genomics, Ludwig Maximilians University of Munich, Munich, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Magdalena Götz
- Institute of Stem Cell Research, GSF-National Research Center for Environment and Health, Neuherberg, Germany.,Munich Cluster for Systems Neurology (SyNergy), Ludwig Maximilians University of Munich, Munich, Germany.,Institute of Physiological Genomics, Ludwig Maximilians University of Munich, Munich, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Axel Imhof
- Protein Analysis Unit (ZfP), Biomedical Center (BMC), Ludwig Maximilians University Munich, Planegg-Martinsried, Germany
| |
Collapse
|
61
|
Hügle M, Lucas X, Weitzel G, Ostrovskyi D, Breit B, Gerhardt S, Einsle O, Günther S, Wohlwend D. 4-Acyl Pyrrole Derivatives Yield Novel Vectors for Designing Inhibitors of the Acetyl-Lysine Recognition Site of BRD4(1). J Med Chem 2016; 59:1518-30. [PMID: 26731611 DOI: 10.1021/acs.jmedchem.5b01267] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Several human diseases, including cancer, show altered signaling pathways resulting from changes in the activity levels of epigenetic modulators. In the past few years, small-molecule inhibitors against specific modulators, including the bromodomain and extra-terminal (BET) bromodomain family of acetylation readers, have shown early promise in the treatment of the genetically defined midline carcinoma and hematopoietic malignancies. We have recently developed a novel potent inhibitor of BET proteins, 1 (XD14[ Angew. Chem., Int. Ed. 2013, 52, 14055]), which exerts a strong inhibitory potential on the proliferation of specific leukemia cell lines. In the study presented here, we designed analogues of 1 to study the potential of substitutions on the 4-acyl pyrrole backbone to occupy additional sites within the substrate recognition site of BRD4(1). The compounds were profiled using ITC, DSF, and X-ray crystallography. We could introduce several substitutions that address previously untargeted areas of the substrate recognition site. This work may substantially contribute to the development of therapeutics with increased target specificity against BRD4-related malignancies.
Collapse
Affiliation(s)
- Martin Hügle
- Institut für Biochemie, Albert-Ludwigs-Universität Freiburg , Albertstrasse 21, D-79104 Freiburg, Germany
| | - Xavier Lucas
- College of Life Sciences, Division of Biological Chemistry and Drug Discovery, University of Dundee , James Black Centre, Dow Street, Dundee, DD1 5EH, United Kingdom
| | - Gerhard Weitzel
- Institut für Organische Chemie, Albert-Ludwigs-Universität Freiburg , Albertstrasse 21, D-79104 Freiburg, Germany
| | - Dmytro Ostrovskyi
- Institut für Organische Chemie, Albert-Ludwigs-Universität Freiburg , Albertstrasse 21, D-79104 Freiburg, Germany
| | - Bernhard Breit
- Institut für Organische Chemie, Albert-Ludwigs-Universität Freiburg , Albertstrasse 21, D-79104 Freiburg, Germany
| | - Stefan Gerhardt
- Institut für Biochemie, Albert-Ludwigs-Universität Freiburg , Albertstrasse 21, D-79104 Freiburg, Germany
| | - Oliver Einsle
- Institut für Biochemie, Albert-Ludwigs-Universität Freiburg , Albertstrasse 21, D-79104 Freiburg, Germany
| | - Stefan Günther
- Institut für Pharmazeutische Wissenschaften, Albert-Ludwigs-Universität Freiburg , Hermann-Herder-Strasse 9, D-79104 Freiburg, Germany
| | - Daniel Wohlwend
- Institut für Biochemie, Albert-Ludwigs-Universität Freiburg , Albertstrasse 21, D-79104 Freiburg, Germany
| |
Collapse
|
62
|
The Functional Analysis of Histone Acetyltransferase MOF in Tumorigenesis. Int J Mol Sci 2016; 17:ijms17010099. [PMID: 26784169 PMCID: PMC4730341 DOI: 10.3390/ijms17010099] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 12/28/2015] [Accepted: 01/05/2016] [Indexed: 12/13/2022] Open
Abstract
Changes in chromatin structure and heritably regulating the gene expression by epigenetic mechanisms, such as histone post-translational modification, are involved in most cellular biological processes. Thus, abnormal regulation of epigenetics is implicated in the occurrence of various diseases, including cancer. Human MOF (males absent on the first) is a member of the MYST (Moz-Ybf2/Sas3-Sas2-Tip60) family of histone acetyltransferases (HATs). As a catalytic subunit, MOF can form at least two distinct multiprotein complexes (MSL and NSL) in human cells. Both complexes can acetylate histone H4 at lysine 16 (H4K16); however, the NSL complex possesses broader substrate specificity and can also acetylate histone H4 at lysines 5 and 8 (H4K5 and H4K8), suggesting the complexity of the intracellular functions of MOF. Silencing of MOF in cells leads to genomic instability, inactivation of gene transcription, defective DNA damage repair and early embryonic lethality. Unbalanced MOF expression and its corresponding acetylation of H4K16 have been found in certain primary cancer tissues, including breast cancer, medulloblastoma, ovarian cancer, renal cell carcinoma, colorectal carcinoma, gastric cancer, as well as non-small cell lung cancer. In this review, we provide a brief overview of MOF and its corresponding histone acetylation, introduce recent research findings that link MOF functions to tumorigenesis and speculate on the potential role that may be relevant to tumorigenic pathways.
Collapse
|
63
|
Shanle EK, Tsun IK, Strahl BD. A course-based undergraduate research experience investigating p300 bromodomain mutations. BIOCHEMISTRY AND MOLECULAR BIOLOGY EDUCATION : A BIMONTHLY PUBLICATION OF THE INTERNATIONAL UNION OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 44:68-74. [PMID: 26537758 PMCID: PMC4982466 DOI: 10.1002/bmb.20927] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/15/2015] [Accepted: 09/29/2015] [Indexed: 05/31/2023]
Abstract
Course-based undergraduate research experiences (CUREs) provide an opportunity for students to engage in experiments with outcomes that are unknown to both the instructor and students. These experiences allow students and instructors to collaboratively bridge the research laboratory and classroom, and provide research experiences for a large number of students relative to traditional individual mentored research. Here, we describe a molecular biology CURE investigating the impact of clinically relevant mutations found in the bromodomain of the p300 transcriptional regulator on acetylated histone interaction. In the CURE, students identified missense mutations in the p300 bromodomain using the Catalogue of Somatic Mutations in Cancer (COSMIC) database and hypothesized the effects of the mutation on the acetyl-binding function of the domain. They cloned and purified the mutated bromodomain and performed peptide pulldown assays to define its potential to bind to acetylated histones. Upon completion of the course, students showed increased confidence performing molecular techniques and reported positively on doing a research project in class. In addition, results generated in the classroom were further validated in the research laboratory setting thereby providing a new model for faculty to engage in both course-based and individual undergraduate research experiences.
Collapse
Affiliation(s)
- Erin K. Shanle
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Biology, University of North Carolina at Pembroke, Pembroke, North Carolina
| | - Ian K. Tsun
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Brian D. Strahl
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
64
|
Yang D, Liang T, Gu Y, Zhao Y, Shi Y, Zuo X, Cao Q, Yang Y, Kan Q. Protein N-arginine methyltransferase 5 promotes the tumor progression and radioresistance of nasopharyngeal carcinoma. Oncol Rep 2015; 35:1703-10. [PMID: 26708443 DOI: 10.3892/or.2015.4513] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/12/2015] [Indexed: 11/06/2022] Open
Abstract
Radiotherapy resistance is the main cause of the the poor prognosis of some nasopharyngeal carcinoma (NPC) patients. Yet, the exact mechanism is still elusive. In the present study, we explored the clinical and biological role of protein arginine methyltransferase 5 (PRMT5) in NPC. Our results revealed that PRMT5 was overexpressed in NPC tissues when compared with that in adjacent non-tumor tissues by quantitative RT-PCR and immunoblotting. High expression of PRMT5 was correlated with adverse outcomes of NPC patients as determined by the scoring of a tissue microarray. Silencing of PRMT5 promoted the radiosensitivity of 5-8F and CNE2 cells as determined by cell proliferation and colony formation assays. Furthermore, fibroblast growth factor receptor 3 (FGFR3) was identified as one of the downstream targets of PRMT5, and the silencing of PRMT5 decreased the mRNA and protein levels of FGFR3 in the 5-8F and CNE2 cells. Silencing of FGFR3 induced similar phenotypes as the inhibition of PRMT5, and re-expression of FGFR3 in 5-8F/shPRMT5 and CNE2/shPRMT5 cells restored the proliferation and colony formation ability induced by irradiation exposure. Our results indicate that PRMT5 is a marker of poor prognosis in NPC patients. PRMT5 promoted the radioresistance of NPC cells via targeting FGFR3, at least partly if not totally. PRMT5 and its downstream effector FGFR3 may be potential targets for anticancer strategy.
Collapse
Affiliation(s)
- Daoke Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University School of Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Tiansong Liang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University School of Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Yue Gu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University School of Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Yulin Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University School of Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Yonggang Shi
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University School of Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Xiaoxiao Zuo
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University School of Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Qinchen Cao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University School of Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Ya Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University School of Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Quancheng Kan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University School of Medicine, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
65
|
Histone Methylation Marks on Circulating Nucleosomes as Novel Blood-Based Biomarker in Colorectal Cancer. Int J Mol Sci 2015; 16:29654-62. [PMID: 26690425 PMCID: PMC4691123 DOI: 10.3390/ijms161226180] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/09/2015] [Accepted: 11/24/2015] [Indexed: 12/15/2022] Open
Abstract
Circulating nucleic acids (CNAs) are under investigation as a liquid biopsy in cancer as potential non-invasive biomarkers, as stable structure in circulation nucleosomes could be valuable sources for detection of cancer-specific alterations in histone modifications. Our interest is in histone methylation marks with a focus on colorectal cancer, one of the leading cancers respective the incidence and mortality. Our previous work included the analysis of trimethylations of lysine 9 on histone 3 (H3K9me3) and of lysine 20 on histone 4 (H4K20me3) by chromatin immuno- precipitation-related PCR in circulating nucleosomes. Here we asked whether global immunologic measurement of histone marks in circulation could be a suitable approach to show their potential as biomarkers. In addition to H3K9me3 and H4K20me3 we also measured H3K27me3 in plasma samples from CRC patients (n = 63) and cancer free individuals (n = 40) by ELISA-based methylation assays. Our results show that of three marks, the amounts of H3K27me3 (p = 0.04) and H4K20me3 (p < 0.001) were significantly lower in CRC patients than in healthy controls. For H3K9me3 similar amounts were measured in both groups. Areas under the curve (AUC) in receiver operating characteristic (ROC) curves indicating the power of CRC detection were 0.620 for H3K27me3, 0.715 for H4K20me3 and 0.769 for the combination of both markers. In conclusion, findings of this preliminary study reveal the potential of blood-based detection of CRC by quantification of histone methylation marks and the additive effect of the marker combination.
Collapse
|
66
|
Inagaki Y, Shiraki K, Sugimoto K, Yada T, Tameda M, Ogura S, Yamamoto N, Takei Y, Ito M. Epigenetic regulation of proliferation and invasion in hepatocellular carcinoma cells by CBP/p300 histone acetyltransferase activity. Int J Oncol 2015; 48:533-40. [PMID: 26676548 DOI: 10.3892/ijo.2015.3288] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 10/23/2015] [Indexed: 11/06/2022] Open
Abstract
Altered epigenetic control of gene expression plays a substantial role in tumor development and progression. Accumulating studies suggest that somatic mutations of CREB binding proteins (CBP)/p300 occur in some cancer cells. CBP/p300 possess histone acetyltransferase (HAT) activity, and are involved in many cellular processes. In this study, we investigated the expression and functional role of CBP/p300 in hepatocellular carcinoma (HCC) using the specific inhibitor C646 of CBP/p300 HAT activity. We examined its effect on several apoptosis-related proteins and invasion-related genes. The results showed that CBP/p300 were highly expressed in HCC tissues and that expression of p300, but not of CBP, was strongly correlated with the malignant character of HCC. C646 inhibited proliferation of HCC cell lines in a dose dependent manner. C646 significantly augmented TRAIL-induced apoptotic sensitivity, which was accompanied by reduced levels of survivin, in HepG2, HLE and SK-HEP1 cells. C646 significantly inhibited invasion of Huh7, HLE and SK-HEP1 cells. The level of matrix metallopeptidase 15 (MMP15) mRNA expression was significantly reduced, whereas the level of laminin alpha 3 (LAMA3) and secreted phosphoprotein 1 (SPP1) mRNA expression was significantly increased in Huh7 cells following exposure to C646. In conclusion, our results suggest that CBP/p300 HAT activity has an important role in malignant transformation, proliferation, apoptotic sensitivity and invasion in HCC. CBP/p300 could be a promising therapeutic target in HCC.
Collapse
Affiliation(s)
- Yuji Inagaki
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Katsuya Shiraki
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Kazushi Sugimoto
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Takazumi Yada
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Masahiko Tameda
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Suguru Ogura
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Norihiko Yamamoto
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yoshiyuki Takei
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Masaaki Ito
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| |
Collapse
|
67
|
Aurora-B and HDAC synergistically regulate survival and proliferation of lymphoma cell via AKT, mTOR and Notch pathways. Eur J Pharmacol 2015; 779:1-7. [PMID: 26638998 DOI: 10.1016/j.ejphar.2015.11.049] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 11/24/2015] [Accepted: 11/26/2015] [Indexed: 11/22/2022]
Abstract
Aurora-B is a protein kinase that functions mainly in the attachment of the mitotic spindle to the centromere. Overexpression of Aurora-B causes unequal distribution of genetic information, creating aneuploidy cells, a hallmark of cancer. Histone deacetylases (HDACs) are a class of enzymes that remove acetyl groups from a ε-N-acetyl lysine amino acid on a histone, allowing the histones to wrap the DNA more tightly, thus globally regulating gene transcription. Additionally, these HDACs can also modify non-histone proteins. Inhibition of HDACs is a potent strategy for cancer treatment. Here, we report that inhibition of Aurora-B and HDAC exerts similar tumor suppressive effects in cells. Knockdown of Aurora-B or inhibition of HDAC achieved the same effect on repression of cell proliferation. Furthermore, we found that the tumor suppressive effect of Aurora-B and HDAC inhibition is due to the induction of cell cycle arrest and/or apoptosis. Mechanistically, we demonstrated that Aurora-B and HDAC can cooperatively regulate AKT, mTOR and Notch pathways.
Collapse
|
68
|
Huang J, Xie Y, Sun X, Zeh HJ, Kang R, Lotze MT, Tang D. DAMPs, ageing, and cancer: The 'DAMP Hypothesis'. Ageing Res Rev 2015; 24:3-16. [PMID: 25446804 DOI: 10.1016/j.arr.2014.10.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 09/24/2014] [Accepted: 10/22/2014] [Indexed: 12/25/2022]
Abstract
Ageing is a complex and multifactorial process characterized by the accumulation of many forms of damage at the molecular, cellular, and tissue level with advancing age. Ageing increases the risk of the onset of chronic inflammation-associated diseases such as cancer, diabetes, stroke, and neurodegenerative disease. In particular, ageing and cancer share some common origins and hallmarks such as genomic instability, epigenetic alteration, aberrant telomeres, inflammation and immune injury, reprogrammed metabolism, and degradation system impairment (including within the ubiquitin-proteasome system and the autophagic machinery). Recent advances indicate that damage-associated molecular pattern molecules (DAMPs) such as high mobility group box 1, histones, S100, and heat shock proteins play location-dependent roles inside and outside the cell. These provide interaction platforms at molecular levels linked to common hallmarks of ageing and cancer. They can act as inducers, sensors, and mediators of stress through individual plasma membrane receptors, intracellular recognition receptors (e.g., advanced glycosylation end product-specific receptors, AIM2-like receptors, RIG-I-like receptors, and NOD1-like receptors, and toll-like receptors), or following endocytic uptake. Thus, the DAMP Hypothesis is novel and complements other theories that explain the features of ageing. DAMPs represent ideal biomarkers of ageing and provide an attractive target for interventions in ageing and age-associated diseases.
Collapse
|
69
|
Lu W, Liu S, Li B, Xie Y, Adhiambo C, Yang Q, Ballard BR, Nakayama KI, Matusik RJ, Chen Z. SKP2 inactivation suppresses prostate tumorigenesis by mediating JARID1B ubiquitination. Oncotarget 2015; 6:771-88. [PMID: 25596733 PMCID: PMC4359254 DOI: 10.18632/oncotarget.2718] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/11/2014] [Indexed: 12/21/2022] Open
Abstract
Aberrant elevation of JARID1B and histone H3 lysine 4 trimethylation (H3K4me3) is frequently observed in many diseases including prostate cancer (PCa), yet the mechanisms on the regulation of JARID1B and H3K4me3 through epigenetic alterations still remain poorly understood. Here we report that Skp2 modulates JARID1B and H3K4me3 levels in vitro in cultured cells and in vivo in mouse models. We demonstrated that Skp2 inactivation decreased H3K4me3 levels, along with a reduction of cell growth, cell migration and malignant transformation of Pten/Trp53 double null MEFs, and further restrained prostate tumorigenesis of Pten/Trp53 mutant mice. Mechanistically, Skp2 decreased the K63-linked ubiquitination of JARID1B by E3 ubiquitin ligase TRAF6, thus decreasing JARID1B demethylase activity and in turn increasing H3K4me3. In agreement, Skp2 deficiency resulted in an increase of JARID1B ubiquitination and in turn a reduction of H3K4me3, and induced senescence through JARID1B accumulation in nucleoli of PCa cells and prostate tumors of mice. Furthermore, we showed that the elevations of Skp2 and H3K4me3 contributed to castration-resistant prostate cancer (CRPC) in mice, and were positively correlated in human PCa specimens. Taken together, our findings reveal a novel network of SKP2-JARID1B, and targeting SKP2 and JARID1B may be a potential strategy for PCa control.
Collapse
Affiliation(s)
- Wenfu Lu
- Department of Biochemistry and Cancer Biology, Meharry Medical College, TN 37208, USA
| | - Shenji Liu
- Department of Biochemistry and Cancer Biology, Meharry Medical College, TN 37208, USA
| | - Bo Li
- Department of Biochemistry and Cancer Biology, Meharry Medical College, TN 37208, USA
| | - Yingqiu Xie
- Department of Biochemistry and Cancer Biology, Meharry Medical College, TN 37208, USA
| | - Christine Adhiambo
- Department of Biochemistry and Cancer Biology, Meharry Medical College, TN 37208, USA
| | - Qing Yang
- Department of Biochemistry and Cancer Biology, Meharry Medical College, TN 37208, USA
| | - Billy R Ballard
- Department of Pathology, Anatomy and Cell Biology, Meharry Medical College, TN 37208, USA
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Robert J Matusik
- Department of Urologic Surgery, Vanderbilt University School of Medicine, TN 37232, USA
| | - Zhenbang Chen
- Department of Biochemistry and Cancer Biology, Meharry Medical College, TN 37208, USA
| |
Collapse
|
70
|
Liu T, Guo Q, Guo H, Hou S, Li J, Wang H. Quantitative analysis of histone H3 and H4 post-translational modifications in doxorubicin-resistant leukemia cells. Biomed Chromatogr 2015; 30:638-44. [DOI: 10.1002/bmc.3608] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 06/26/2015] [Accepted: 08/24/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Tao Liu
- International Joint Cancer Institute; Second Military Medical University; Shanghai China
| | - Qingcheng Guo
- International Joint Cancer Institute; Second Military Medical University; Shanghai China
- State Key Laboratory of Antibody Medicine and Targeted Therapy; Shanghai Key Laboratory of Cell Engineering; Shanghai China
| | - Huaizu Guo
- International Joint Cancer Institute; Second Military Medical University; Shanghai China
- State Key Laboratory of Antibody Medicine and Targeted Therapy; Shanghai Key Laboratory of Cell Engineering; Shanghai China
| | - Sheng Hou
- International Joint Cancer Institute; Second Military Medical University; Shanghai China
- PLA General Hospital Cancer Center; PLA Postgraduate School of Medicine; Beijing China
| | - Jing Li
- International Joint Cancer Institute; Second Military Medical University; Shanghai China
| | - Hao Wang
- International Joint Cancer Institute; Second Military Medical University; Shanghai China
- PLA General Hospital Cancer Center; PLA Postgraduate School of Medicine; Beijing China
| |
Collapse
|
71
|
Mir AR, Moinuddin. Glycoxidation of histone proteins in autoimmune disorders. Clin Chim Acta 2015; 450:25-30. [DOI: 10.1016/j.cca.2015.07.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/26/2015] [Accepted: 07/28/2015] [Indexed: 01/08/2023]
|
72
|
|
73
|
Singh R, Harshman SW, Ruppert AS, Mortazavi A, Lucas DM, Thomas-Ahner JM, Clinton SK, Byrd JC, Freitas MA, Parthun MR. Proteomic profiling identifies specific histone species associated with leukemic and cancer cells. Clin Proteomics 2015; 12:22. [PMID: 26321891 PMCID: PMC4551702 DOI: 10.1186/s12014-015-9095-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 08/12/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chromatin is an extraordinarily complex structure. Much of this complexity results from the presence of numerous histone post-translational modifications and histone variants. Alterations in the patterns of histone post-translational modifications are emerging as a feature of many types of cancer and have been shown to have prognostic value. RESULTS We have applied a liquid chromatography/mass spectrometry-based approach to comprehensively characterize the histone proteome in primary samples from chronic lymphocytic leukemia (CLL) patients, as well as bladder and breast cancer cell culture models. When compared to non-malignant CD19+ B cells from healthy donors, the CLL histone proteome showed a distinct signature of differentially expressed species, spanning all the histones studied and including both post-translationally modified species and unmodified, non-allelic replication-dependent histone isoforms. However, the large changes in histone H3 and H4 that are characteristic of many cancer types were not observed. One of species of H2A (mass = 14,063 Da) was the most strongly associated with time to treatment in CLL patients. CLL patient samples also demonstrated histone profiles that were distinct from those of the bladder and breast cancer cells. CONCLUSIONS Signatures of histone profiles are complex and can distinguish between healthy individuals and CLL patients and may provide prognostic markers. In addition, histone profiles may define tissue specific malignancies.
Collapse
Affiliation(s)
- Rajbir Singh
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210 USA
| | - Sean W Harshman
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210 USA
| | - Amy S Ruppert
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - Amir Mortazavi
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - David M Lucas
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210 USA ; Division of Medicinal Chemistry in the College of Pharmacy, The Ohio State University, Columbus, OH 43210 USA
| | | | - Steven K Clinton
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - John C Byrd
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - Michael A Freitas
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210 USA
| | - Mark R Parthun
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
74
|
Maleszewska M, Kaminska B. Deregulation of histone-modifying enzymes and chromatin structure modifiers contributes to glioma development. Future Oncol 2015; 11:2587-601. [PMID: 26289459 DOI: 10.2217/fon.15.171] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The epigenetic landscape is deregulated in cancer due to aberrant activation or inactivation of enzymes maintaining and modifying the epigenome. Histone modifications and global aberrations at the histone level may result in distorted patterns of gene expression, and malfunction of proteins that regulate chromatin modification and remodeling. Recent whole genome studies demonstrated that histones and chaperone proteins harbor mutations that may result in gross alterations of the epigenome leading to genome instability. Glioma development is a multistep process, involving genetic and epigenetic alterations. This review summarizes newly identified mechanisms affecting expression/functions of histone-modifying enzymes and chromatin modifiers in gliomas. We discuss recent approaches to overcome epigenetic alterations with histone-modifying enzyme inhibitors and their prospects for glioma therapy.
Collapse
Affiliation(s)
- Marta Maleszewska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, 3 Pasteur Str., 02-093 Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, 3 Pasteur Str., 02-093 Warsaw, Poland
| |
Collapse
|
75
|
Abstract
The current healthcare system is focused on disease management. Our current approach to treatment begins only after the diagnosis, and then attempts to treat the symptoms and prevent the progression. Despite increased global healthcare spending, there has been an increasing incidence, and severity of diseases pointing to impaired health of the populace. This progressive deterioration in general health has created an unsustainable increase in healthcare costs that has hampered the economy. Much of the rising costs in healthcare are secondary to treating the progression of preventable diseases and focus on creating new treatments. There has been an ongoing discussion of incorporating a “defense” or prevention as part of our health system. However, there are few established guidelines beyond tactical use of vaccination in known infectious diseases and screening for chronic diseases and cancers. Ayurveda has the core competency and strategy for prevention of disease. Sushruta has propounded the laws of health, which are unknown to the current healthcare system. This article describes these laws and strategic combination of Ayurveda (defense) and modern medicine (offense) to create a complete healthcare system. This system is called Symbiohealth and is potentially more effective, less expensive, less toxic and creates a healthier society.
Collapse
Affiliation(s)
- Gopal Basisht
- Senior Consultant Rheumatology, Orlando Health, Orlando, Florida, USA
| |
Collapse
|
76
|
Gu R, Liu T, Zhu X, Gan H, Wu Z, Li J, Zheng Y, Dou G, Meng Z. Development and validation of a sensitive HPLC-MS/MS method for determination of chidamide (epidaza), a new benzamide class of selective histone deacetylase inhibitor, in human plasma and its clinical application. J Chromatogr B Analyt Technol Biomed Life Sci 2015; 1000:181-6. [PMID: 26245362 DOI: 10.1016/j.jchromb.2015.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 06/29/2015] [Accepted: 07/02/2015] [Indexed: 01/22/2023]
Abstract
Chidamide (epidaza), a new oral isotype-selective histone deacetylase inhibitor (HDACi), which is just approved in China for the treatment of recurrent or refractory peripheral T-cell lymphoma (PTCL) in December 2014, is the first listed benzamide class of HDACi in the world, and is currently undergoing global clinical trials for solid tumor treatments. Here, we report a sensitive, rapid and robust HPLC-MS/MS method for determination of chidamide in human plasma. Plasma sample was subjected to a simple acetonitrile protein precipitation containing MS-275 used as an internal standard (IS). Chromatography was performed on a Hypersil GOLD C18 analytical column, using a gradient methanol/water mobile phase containing 0.1% formic acid. A tandem mass spectrometer equipped with electrospray ionization source was used as detector and operated in the positive-ion mode. Selected reaction monitoring (SRM) using the precursor/ product transitions (m/z) of 391.1/265.1 for chidamide and 377.1/359.2 for IS were used for quantification, respectively. Good linearity was obtained in the range of 1-1000ng/mL. The method gave R.S.D.% values for precision always lower than 13.8% and R.E.% values for accuracy between -3.7 and 9.1%. In addition, the specificity, recovery, stability and matrix effect were satisfactory too. The method is now being successfully applied to plasma samples as part of an ongoing chidamide phase Ib clinical trial in patients with solid tumors, and had demonstrated consistent AUClast and t1/2 results with the published phase I pharmacokinetic data, which was also analyzed by this method, thus further confirming the reproducibility and accuracy during its clinical application. Considering the excellent performance of this method, it will continue being utilized for future clinical developments of chidamide and for routine monitoring of plasma exposure of chidamide during its clinical therapy.
Collapse
Affiliation(s)
- Ruolan Gu
- State Key Laboratory of Drug Metabolism and Pharmacokinetics, Laboratory of Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Haidian District, Beijing 100850, China
| | - Taoyun Liu
- State Key Laboratory of Drug Metabolism and Pharmacokinetics, Laboratory of Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Haidian District, Beijing 100850, China
| | - Xiaoxia Zhu
- State Key Laboratory of Drug Metabolism and Pharmacokinetics, Laboratory of Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Haidian District, Beijing 100850, China
| | - Hui Gan
- State Key Laboratory of Drug Metabolism and Pharmacokinetics, Laboratory of Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Haidian District, Beijing 100850, China
| | - Zhuona Wu
- State Key Laboratory of Drug Metabolism and Pharmacokinetics, Laboratory of Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Haidian District, Beijing 100850, China
| | - Jian Li
- State Key Laboratory of Drug Metabolism and Pharmacokinetics, Laboratory of Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Haidian District, Beijing 100850, China
| | - Ying Zheng
- State Key Laboratory of Drug Metabolism and Pharmacokinetics, Laboratory of Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Haidian District, Beijing 100850, China
| | - Guifang Dou
- State Key Laboratory of Drug Metabolism and Pharmacokinetics, Laboratory of Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Haidian District, Beijing 100850, China.
| | - Zhiyun Meng
- State Key Laboratory of Drug Metabolism and Pharmacokinetics, Laboratory of Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Haidian District, Beijing 100850, China.
| |
Collapse
|
77
|
Dynamic epigenetic regulation of glioblastoma tumorigenicity through LSD1 modulation of MYC expression. Proc Natl Acad Sci U S A 2015; 112:E4055-64. [PMID: 26159421 DOI: 10.1073/pnas.1501967112] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The available evidence suggests that the lethality of glioblastoma is driven by small subpopulations of cells that self-renew and exhibit tumorigenicity. It remains unclear whether tumorigenicity exists as a static property of a few cells or as a dynamically acquired property. We used tumor-sphere and xenograft formation as assays for tumorigenicity and examined subclones isolated from established and primary glioblastoma lines. Our results indicate that glioblastoma tumorigenicity is largely deterministic, yet the property can be acquired spontaneously at low frequencies. Further, these dynamic transitions are governed by epigenetic reprogramming through the lysine-specific demethylase 1 (LSD1). LSD depletion increases trimethylation of histone 3 lysine 4 at the avian myelocytomatosis viral oncogene homolog (MYC) locus, which elevates MYC expression. MYC, in turn, regulates oligodendrocyte lineage transcription factor 2 (OLIG2), SRY (sex determining region Y)-box 2 (SOX2), and POU class 3 homeobox 2 (POU3F2), a core set of transcription factors required for reprogramming glioblastoma cells into stem-like states. Our model suggests epigenetic regulation of key transcription factors governs transitions between tumorigenic states and provides a framework for glioblastoma therapeutic development.
Collapse
|
78
|
Morales La Madrid A, Hashizume R, Kieran MW. Future Clinical Trials in DIPG: Bringing Epigenetics to the Clinic. Front Oncol 2015; 5:148. [PMID: 26191506 PMCID: PMC4486770 DOI: 10.3389/fonc.2015.00148] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/16/2015] [Indexed: 11/18/2022] Open
Abstract
In spite of major recent advances in diffuse intrinsic pontine glioma (DIPG) molecular characterization, this body of knowledge has not yet translated into better treatments. To date, more than 250 clinical trials evaluating radiotherapy along with conventional cytotoxic chemotherapy as well as newer biologic agents have failed to improve the dismal outcome when compared to palliative radiation alone. The biology of DIPG remained unknown until recently when the neurosurgical expertise along with the recognition by the scientific and clinical community of the importance of tissue sampling at diagnosis; ideally, in the context of a clinical trial and by trained neurosurgical teams to maximize patient safety. These pre-treatment tumor samples, and others coming from tissue obtained post-mortem, have yielded new insights into DIPG molecular pathogenesis. We now know that DIPG comprises a heterogeneous disease with variable molecular phenotypes, different from adult high-grade glioma, other non-pontine pediatric high-grade gliomas, and even between pontine gliomas. The discovery of histone H3.3 or H3.1 mutations has been an important step forward in understanding tumor formation, maintenance, and progression. Pharmacologic reversal of DIPG histone demethylation therefore offers an important potential intervention strategy for the treatment of DIPG. To date, clinical trials of newly diagnosed or progressive DIPG with epigenetic (histone) modifiers have been unsuccessful. Whether this failure represents limited activity of the agents used, their CNS penetration, redundant pathways within the tumor, or the possibility that histone mutations are necessary only to initiate DIPGs but not maintain their growth, suggest that a great deal still needs to be elucidated in both the underlying biology of these pathways and the drugs designed to target them. In this review, we will discuss the role of both epigenetic and genetic mutations within DIPG and the development of treatment strategies directed against the unique abnormalities present in this disease.
Collapse
Affiliation(s)
- Andres Morales La Madrid
- Pediatric Neuro-Oncology, Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Deu , Barcelona , Spain
| | - Rintaro Hashizume
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University , Chicago, IL , USA ; Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University , Chicago, IL , USA
| | - Mark W Kieran
- Pediatric Neuro-Oncology, Department of Pediatric Hematology/Oncology, Dana-Farber Cancer Institute, Boston Children's Hospital , Boston, MA , USA
| |
Collapse
|
79
|
Epigenetics of Meningiomas. BIOMED RESEARCH INTERNATIONAL 2015; 2015:532451. [PMID: 26101774 PMCID: PMC4458517 DOI: 10.1155/2015/532451] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 12/14/2014] [Indexed: 12/20/2022]
Abstract
Meningiomas account for one-third of all adult central nervous system tumours and are divided into three WHO grades. In contrast to the relatively well characterized genetic alterations, our current understanding of epigenetic modifications involved in the meningioma-genesis and progression is rather incomplete. Contrary to genetic alterations, epigenetic changes do not alter the primary DNA sequence and their reversible nature serves as an excellent basis for prevention and development of novel personalised tumour therapies. Indeed, growing body of evidence suggests that disturbed epigenetic regulation plays a key role in the pathogenesis of meningiomas. Altered DNA methylation, microRNA expression, histone, and chromatin modifications are frequently noted in meningiomas bearing prognostic and therapeutic relevance. In this review we provide an overview on recently identified epigenetic alterations in meningiomas and discuss their role in tumour initiation, progression, and recurrence.
Collapse
|
80
|
Abstract
Genetic and epigenetic changes are at the root of all cancers. The epigenetic component involves alterations of the post-synthetic modifications of DNA (methylation) and histones (histone posttranslational modifications, PTMs) as well as of those of their molecular "writers," "readers," and "erasers." Noncoding RNAs (ncRNA) can also play a role. Here, we focus on the involvement of histone alterations in cancer, in particular that of the histone variant H2A.Z in the etiology of prostate cancer. The structural mechanisms putatively responsible for the contribution of H2A.Z to oncogenic gene expression programs are first described, followed by what is currently known about the involvement of this histone variant in the regulation of androgen receptor regulated gene expression. The implications of this and their relevance to oncogene deregulation in different stages of prostate cancer, including the progression toward androgen independence, are discussed. This review underscores the increasing awareness of the epigenetic contribution of histone variants to oncogenic progression.
Collapse
Affiliation(s)
- Deanna Dryhurst
- Department of Biochemistry and Microbiology, University of Victoria, Petch building, 258a, Victoria, British Columbia Canada V8W 3P6
- ImmunoPrecise Antibodies Ltd., 3204-4464 Markham St., Victoria, British Columbia Canada V8Z 7X8
| | - Juan Ausió
- Department of Biochemistry and Microbiology, University of Victoria, Petch building, 258a, Victoria, British Columbia Canada V8W 3P6
| |
Collapse
|
81
|
Guo X, Tian Z, Wang X, Pan S, Huang W, Shen Y, Gui Y, Duan X, Cai Z. Regulation of histone demethylase KDM6B by hypoxia-inducible factor-2α. Acta Biochim Biophys Sin (Shanghai) 2015; 47:106-13. [PMID: 25520177 DOI: 10.1093/abbs/gmu122] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Lysine (K)-specific demethylase 6B (KDM6B) is a histone H3K27 demethylase, which specifically catalyzes the demethylation of H3 lysine-27 tri/dimethylation (H3K27me3/2). KDM6B can activate gene transcription by promoting transcriptional elongation which is associated with RNA polymerase II and related elongation factors. So KDM6B is important for the regulation of gene expression. Previous studies have indicated that several histone demethylases such as KDM3A, KDM4B, and KDM4C are regulated by hypoxia-inducible factor (HIF). But, the effect of hypoxia on KDM6B is not fully understood. In this study, we found that the expression levels of KDM6B mRNA and protein are modestly up-regulated under hypoxia (1% O2) or mimic hypoxia (desferrioxamine mesylate or CoCl2 treatment) (P<0.05). The result of RNAi shows that the up-regulation of KDM6B is dependent on HIF-2α, but not on HIF-1α. The result of chromatin immunoprecipitation assay indicates that there is a hypoxia response element in KDM6B promoter (-4041 to -4037). The result of Co-IP assay indicates that KDM6B can form complex with HIF-2α or HIF-1α. The knockdown experiment implies that KDM6B is a potential regulator for HIF-2α target genes. These data demonstrate that KDM6B is a new hypoxia response gene regulated by HIF-2α. Our results also show that KDM6B is a potential co-activator of HIF-α, which is important for the activation of hypoxia response genes.
Collapse
Affiliation(s)
- Xiaoqiang Guo
- Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Zhantao Tian
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Xuliang Wang
- Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen 518036, China
| | - Shuhong Pan
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Weiren Huang
- Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Yongqing Shen
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Yaoting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen 518036, China
| | - Xianglin Duan
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Zhiming Cai
- Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| |
Collapse
|
82
|
Park KC, Heo JH, Jeon JY, Choi HJ, Jo AR, Kim SW, Kwon HJ, Hong SJ, Han KS. The novel histone deacetylase inhibitor, N-hydroxy-7-(2-naphthylthio) hepatonomide, exhibits potent antitumor activity due to cytochrome-c-release-mediated apoptosis in renal cell carcinoma cells. BMC Cancer 2015; 15:19. [PMID: 25613585 PMCID: PMC4318161 DOI: 10.1186/s12885-014-1003-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2014] [Indexed: 12/25/2022] Open
Abstract
Background Epigenetic modifications play a critical role in the regulation of all DNA-based processes, such as transcription, repair, and replication. Inappropriate histone modifications can result in dysregulation of cell growth, leading to neoplastic transformation and cell death. Renal tumors have been shown to have a higher global methylation percentage and reduced histone acetylation. Preclinical models have revealed that histone gene modifiers and epigenetic alterations play important roles in renal cell carcinoma (RCC) tumorigenesis. Recently, a novel HDAC inhibitor, N-hydroxy-7-(2-naphthylthio) heptanomide (HNHA), has been introduced as an example of a new class of anti-cancer agents. The anti-cancer activity of HNHA and the underlying mechanisms of action remain to be clarified. Methods The MTS assay using a panel of RCC cells was used to evaluate the anti-proliferative effects of HNHA. The established HDAC inhibitors, SAHA and TSA, were used for comparison. Western blotting analysis was performed to investigate the acetylation of histone H3 and the expression of apoptotic markers in vitro and in vivo. Subcellular fractionation was performed to evaluate expression of Bax and cytochrome c in the cytosol and mitochondria, and also translocation of cytochrome c from the cytoplasm to the nucleus. A confocal microscopic evaluation was performed to confirm inhibition of cell proliferation, induction of apoptosis, and the nuclear translocation of cytochrome c in RCC cells. Results In this study, we investigated the apoptosis-inducing activity of HNHA in cultured kidney cancer cells. Apoptosis in the HNHA-treated group was induced significantly, with marked caspase activation and Bcl-2 suppression in RCC cells in vitro and in vivo. HNHA treatment caused cytochrome c release from mitochondria, which was mediated by increased Bax expression and caspase activation. HNHA also induced nuclear translocation of cytochrome c, suggesting that HNHA can induce caspase-independent nuclear apoptosis in RCC cells. An in vivo study showed that HNHA had greater anti-tumor and pro-apoptotic effects on RCC xenografts than the established HDAC inhibitors. Conclusions HNHA has more potent anti-tumor activity than established HDAC inhibitors. Its activities are mediated by caspase-dependent and cytochrome-c-mediated apoptosis in RCC cells. These results suggest that HNHA may offer a new therapeutic approach to RCC.
Collapse
|
83
|
Thinnes CC, England KS, Kawamura A, Chowdhury R, Schofield CJ, Hopkinson RJ. Targeting histone lysine demethylases - progress, challenges, and the future. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1839:1416-32. [PMID: 24859458 PMCID: PMC4316176 DOI: 10.1016/j.bbagrm.2014.05.009] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 05/06/2014] [Accepted: 05/13/2014] [Indexed: 12/20/2022]
Abstract
N-Methylation of lysine and arginine residues has emerged as a major mechanism of transcriptional regulation in eukaryotes. In humans, N(ε)-methyllysine residue demethylation is catalysed by two distinct subfamilies of demethylases (KDMs), the flavin-dependent KDM1 subfamily and the 2-oxoglutarate- (2OG) dependent JmjC subfamily, which both employ oxidative mechanisms. Modulation of histone methylation status is proposed to be important in epigenetic regulation and has substantial medicinal potential for the treatment of diseases including cancer and genetic disorders. This article provides an introduction to the enzymology of the KDMs and the therapeutic possibilities and challenges associated with targeting them, followed by a review of reported KDM inhibitors and their mechanisms of action from kinetic and structural perspectives.
Collapse
Affiliation(s)
- Cyrille C Thinnes
- The Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | | | - Akane Kawamura
- The Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | | | | | | |
Collapse
|
84
|
Genovese F, Karsdal MA, Leeming DJ, Scholze A, Tepel M. Association of versican turnover with all-cause mortality in patients on haemodialysis. PLoS One 2014; 9:e111134. [PMID: 25354390 PMCID: PMC4212982 DOI: 10.1371/journal.pone.0111134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/22/2014] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Cardiovascular diseases are among the most common causes of mortality in renal failure patients undergoing haemodialysis. A high turnover rate of the proteoglycan versican, represented by the increased presence of its fragmentation products in plasma, has previously been associated with cardiovascular diseases. The objective of the study was to investigate the association of versican turnover assessed in plasma with survival in haemodialysis patients. METHODS A specific matrix metalloproteinase-generated neo-epitope fragment of versican (VCANM) was measured in plasma of 364 haemodialysis patients with a 5-years follow-up, using a robust competitive enzyme-linked immunosorbent assays. Association between VCANM plasma concentration and survival was assessed by Kaplan-Meier analysis and adjusted Cox model. RESULTS Haemodialysis patients with plasma VCANM concentrations in the lowest quartile had increased risk of death (odds ratio, as compared to the highest quartile: 7.1, p<0.001), with a reduced survival of 152 days compared to 1295 days for patients with plasma VCANM in the highest quartile. Multivariate analysis showed that low VCANM (p<0.001) and older age (p<0.001) predicted death in haemodialysis patients. CONCLUSIONS Low concentrations of the versican fragment VCANM in plasma were associated with higher risk of death among haemodialysis patients. A possible protective role for the examined versican fragment is suggested.
Collapse
Affiliation(s)
- Federica Genovese
- Nordic Bioscience, Fibrosis Biology and Biomarkers, Herlev, Denmark
- * E-mail:
| | | | - Diana J. Leeming
- Nordic Bioscience, Fibrosis Biology and Biomarkers, Herlev, Denmark
| | - Alexandra Scholze
- Odense University Hospital, Department of Nephrology, Institute for Molecular Medicine, Cardiovascular and Renal Research, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Martin Tepel
- Odense University Hospital, Department of Nephrology, Institute for Molecular Medicine, Cardiovascular and Renal Research, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
85
|
Kebede AF, Schneider R, Daujat S. Novel types and sites of histone modifications emerge as players in the transcriptional regulation contest. FEBS J 2014; 282:1658-74. [DOI: 10.1111/febs.13047] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/03/2014] [Accepted: 09/09/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Adam F. Kebede
- Institut de Génétique et de Biologie Moléculaire et Cellulaire; CNRS UMR 7104 - Inserm U964; Université de Strasbourg; Illkirch France
| | - Robert Schneider
- Institut de Génétique et de Biologie Moléculaire et Cellulaire; CNRS UMR 7104 - Inserm U964; Université de Strasbourg; Illkirch France
| | - Sylvain Daujat
- Institut de Génétique et de Biologie Moléculaire et Cellulaire; CNRS UMR 7104 - Inserm U964; Université de Strasbourg; Illkirch France
| |
Collapse
|
86
|
Biegel JA, Busse TM, Weissman BE. SWI/SNF chromatin remodeling complexes and cancer. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2014; 166C:350-66. [PMID: 25169151 DOI: 10.1002/ajmg.c.31410] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The identification of mutations and deletions in the SMARCB1 locus in chromosome band 22q11.2 in pediatric rhabdoid tumors provided the first evidence for the involvement of the SWI/SNF chromatin remodeling complex in cancer. Over the last 15 years, alterations in more than 20 members of the complex have been reported in a variety of human tumors. These include germline mutations and copy number alterations in SMARCB1, SMARCA4, SMARCE1, and PBRM1 that predispose carriers to both benign and malignant neoplasms. Somatic mutations, structural abnormalities, or epigenetic modifications that lead to reduced or aberrant expression of complex members have now been reported in more than 20% of malignancies, including both solid tumors and hematologic disorders in both children and adults. In this review, we will highlight the role of SMARCB1 in cancer as a paradigm for other tumors with alterations in SWI/SNF complex members and demonstrate the broad spectrum of mutations observed in complex members in different tumor types.
Collapse
|
87
|
Vianello P, Botrugno OA, Cappa A, Ciossani G, Dessanti P, Mai A, Mattevi A, Meroni G, Minucci S, Thaler F, Tortorici M, Trifiró P, Valente S, Villa M, Varasi M, Mercurio C. Synthesis, biological activity and mechanistic insights of 1-substituted cyclopropylamine derivatives: a novel class of irreversible inhibitors of histone demethylase KDM1A. Eur J Med Chem 2014; 86:352-63. [PMID: 25173853 DOI: 10.1016/j.ejmech.2014.08.068] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/25/2014] [Accepted: 08/26/2014] [Indexed: 11/25/2022]
Abstract
Histone demethylase KDM1A (also known as LSD1) has become an attractive therapeutic target for the treatment of cancer as well as other disorders such as viral infections. We report on the synthesis of compounds derived from the expansion of tranylcypromine as a chemical scaffold for the design of novel demethylase inhibitors. These compounds, which are substituted on the cyclopropyl core moiety, were evaluated for their ability to inhibit KDM1A in vitro as well as to function in cells by modulating the expression of Gfi-1b, a well recognized KDM1A target gene. The molecules were all found to covalently inhibit KDM1A and to become increasingly selective against human monoamine oxidases MAO A and MAO B through the introduction of bulkier substituents on the cyclopropylamine ring. Structural and biochemical analysis of selected trans isomers showed that the two stereoisomers are endowed with similar inhibitory activities against KDM1A, but form different covalent adducts with the FAD co-enzyme.
Collapse
Affiliation(s)
- Paola Vianello
- Drug Discovery Unit, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy.
| | - Oronza A Botrugno
- Drug Discovery Unit, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Anna Cappa
- Drug Discovery Unit, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Giuseppe Ciossani
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 1, 27100 Pavia, Italy
| | - Paola Dessanti
- Drug Discovery Unit, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Antonello Mai
- Pasteur Institute - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, University "La Sapienza", P.le A. Moro 5, 00185 Roma, Italy
| | - Andrea Mattevi
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 1, 27100 Pavia, Italy.
| | - Giuseppe Meroni
- Drug Discovery Unit, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Saverio Minucci
- Drug Discovery Unit, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy; Department of Biosciences, University of Milan, Via Celoria, 26, 20133 Milan, Italy
| | - Florian Thaler
- Drug Discovery Unit, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy.
| | - Marcello Tortorici
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 1, 27100 Pavia, Italy
| | - Paolo Trifiró
- Drug Discovery Unit, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Sergio Valente
- Pasteur Institute - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, University "La Sapienza", P.le A. Moro 5, 00185 Roma, Italy
| | - Manuela Villa
- Drug Discovery Unit, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Mario Varasi
- Drug Discovery Unit, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Ciro Mercurio
- Drug Discovery Unit, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| |
Collapse
|
88
|
Bai Y, Fang N, Gu T, Kang Y, Wu J, Yang D, Zhang H, Suo Z, Ji S. HOXA11 gene is hypermethylation and aberrant expression in gastric cancer. Cancer Cell Int 2014; 14:79. [PMID: 25788862 PMCID: PMC4364045 DOI: 10.1186/s12935-014-0079-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/29/2014] [Indexed: 12/11/2022] Open
Abstract
Background Aberrant DNA methylation is an acquired epigenetic alteration that serves as an alternative to genetic defects in the inactivation of tumor suppressor genes and other genes in diverse human cancers. Gastric carcinoma is one of the tumors with a high frequency of aberrant methylation in promoter region. Hence we investigated the promoter methylation status and expression level of HOXA11 gene which may involve in GC development. Methods Thirty-two surgical excised gastric cancer specimens, twelve paired adjacent non-cancerous specimens and seven normal gastric mucosas were examined. The methylation status and expression level of HOXA11 gene were determined by bisulfite sequencing polymerase chain reaction (BSP), real-time polymerase chain reaction (RT-PCR) and immunohistochemistry (IHC) respectively. HOXA11 expression was knocked-down with siRNA to mimic HOXA11 gene hypermethylation and ability of cell proliferation and migration was determinate. In addition, we analyzed and correlated the findings with clinicopathological features. Results The methylation level of HOXA11 gene in gastric cancer tissues and adjacent non-cancerous tissues were higher than those in normal gastric mucosa (P < 0.05). The methylation level was higher in TNM III and IV patients of GC than those in TNM I and II patients (P < 0.05). The expression of HOXA11 mRNA and protein decreased in normal gastric mucosa, peri-cancer tissue and GC (P < 0.05). HOXA11 expression was inversely correlated with DNA methylation (P < 0.05). Knocked-down of HOXA11 expression with siRNA in BGC-823 cells enhanced cell proliferation compared with control, but no significant different was observed in migration ability. Conclusion Hypermethylation and decreased expression of HOXA11 gene may be involved in the carcinogenesis and development of GC and may provide useful information for the prediction of the malignant behaviors of GC. And the expression of HOXA11 is impaired by DNA methylation. However, repression of HOXA11 expression promoted BGC-823 cell proliferation.
Collapse
Affiliation(s)
- Yinguo Bai
- Department of Gastroenterology, Huaihe Hospital of Henan University, Kaifeng 475000, Henan Province, China ; Department of Biochemistry and Molecular Biology, Medical School of Henan University, Kaifeng 475004, Henan Province, China
| | - Na Fang
- Department of Biochemistry and Molecular Biology, Medical School of Henan University, Kaifeng 475004, Henan Province, China
| | - Tingxun Gu
- Department of Biochemistry and Molecular Biology, Medical School of Henan University, Kaifeng 475004, Henan Province, China
| | - Yuhua Kang
- Department of Gastroenterology, Huaihe Hospital of Henan University, Kaifeng 475000, Henan Province, China
| | - Jiang Wu
- Department of pathology, Huaihe Hospital of Henan University, Kaifeng 475000, Henan Province, China
| | - Desheng Yang
- Department of Gastroenterology, Huaihe Hospital of Henan University, Kaifeng 475000, Henan Province, China
| | - Hui Zhang
- Department of Gastroenterology, Huaihe Hospital of Henan University, Kaifeng 475000, Henan Province, China
| | - Zhimin Suo
- Department of Gastroenterology, Huaihe Hospital of Henan University, Kaifeng 475000, Henan Province, China
| | - Shaoping Ji
- Department of Biochemistry and Molecular Biology, Medical School of Henan University, Kaifeng 475004, Henan Province, China
| |
Collapse
|
89
|
Zane L, Sharma V, Misteli T. Common features of chromatin in aging and cancer: cause or coincidence? Trends Cell Biol 2014; 24:686-94. [PMID: 25103681 DOI: 10.1016/j.tcb.2014.07.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/30/2014] [Accepted: 07/01/2014] [Indexed: 02/06/2023]
Abstract
Age is a major risk factor for cancer. Alterations in DNA methylation, histone modifications, chromatin structure, and epigenetic regulatory mechanisms are prominent hallmarks of both the aging process and cancer. Intriguingly--or possibly coincidentally--several chromatin features are common between aging and cancer. Here we ask whether, and if so how, aging-associated chromatin modifications contribute to tumor susceptibility and tumorigenesis.
Collapse
Affiliation(s)
- Linda Zane
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vivek Sharma
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tom Misteli
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
90
|
Weng MK, Natarajan K, Scholz D, Ivanova VN, Sachinidis A, Hengstler JG, Waldmann T, Leist M. Lineage-specific regulation of epigenetic modifier genes in human liver and brain. PLoS One 2014; 9:e102035. [PMID: 25054330 PMCID: PMC4108363 DOI: 10.1371/journal.pone.0102035] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 06/13/2014] [Indexed: 12/14/2022] Open
Abstract
Despite an abundance of studies on chromatin states and dynamics, there is an astonishing dearth of information on the expression of genes responsible for regulating histone and DNA modifications. We used here a set of 156 defined epigenetic modifier genes (EMG) and profiled their expression pattern in cells of different lineages. As reference value, expression data from human embryonic stem cells (hESC) were used. Hepatocyte-like cells were generated from hESC, and their EMG expression was compared to primary human liver cells. In parallel, we generated postmitotic human neurons (Lu d6), and compared their relative EMG expression to human cortex (Ctx). Clustering analysis of all cell types showed that neuronal lineage samples grouped together (94 similarly regulated EMG), as did liver cells (61 similarly-regulated), while the two lineages were clearly distinct. The general classification was followed by detailed comparison of the major EMG groups; genes that were higher expressed in differentiated cells than in hESC included the acetyltransferase KAT2B and the methyltransferase SETD7. Neuro-specific EMGs were the histone deacetylases HDAC5 and HDAC7, and the arginine-methyltransferase PRMT8. Comparison of young (Lu d6) and more aged (Ctx) neuronal samples suggested a maturation-dependent switch in the expression of functionally homologous proteins. For instance, the ratio of the histone H3 K27 methyltransfereases, EZH1 to EZH2, was high in Ctx and low in Lu d6. The same was observed for the polycomb repressive complex 1 (PRC1) subunits CBX7 and CBX8. A large proportion of EMGs in differentiated cells was very differently expressed than in hESC, and absolute levels were significantly higher in neuronal samples than in hepatic cells. Thus, there seem to be distinct qualitative and quantitative differences in EMG expression between cell lineages.
Collapse
Affiliation(s)
- Matthias K. Weng
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
- * E-mail:
| | - Karthick Natarajan
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne (UKK), Cologne, Germany
| | - Diana Scholz
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Violeta N. Ivanova
- Nycomed-Chair for Bioinformatics and Information Mining, Dept. of Computer and Information Science, University of Konstanz, Konstanz, Germany
| | - Agapios Sachinidis
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne (UKK), Cologne, Germany
| | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, Dortmund, Germany
| | - Tanja Waldmann
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Marcel Leist
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| |
Collapse
|
91
|
Abstract
A major biomedical advance from recent years was the finding that gene expression and phenotypic traits may be shaped by potentially reversible and heritable modifications that occur without altering the sequence of the nucleotides, and became known as epigenetic changes. The term 'epigenetics' dates back to the 1940s, when it was first used in context of cellular differentiation decisions that are made during development. Since then, our understanding of epigenetic modifications that govern development and disease expanded considerably. The contribution of epigenetic changes to shaping phenotypes brings at least two major clinically relevant benefits. One of these, stemming from the reversibility of epigenetic changes, involves the possibility to therapeutically revert epigenetic marks to re-establish prior gene expression patterns. The strength and the potential of this strategy are illustrated by the first four epigenetic drugs that were approved in recent years and by the additional candidates that are at various stages in preclinical studies and clinical trials. The second particularity is the finding that epigenetic changes precede the appearance of histopathological modifications. This has the potential to facilitate the emergence of epigenetic biomarkers, some of which already entered the clinical arena, catalysing a major shift in prophylactic and therapeutic strategies, and promising to fill a decades-old gap in preventive medicine.
Collapse
Affiliation(s)
- R A Stein
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
92
|
Carvalho S, Vítor AC, Sridhara SC, Martins FB, Raposo AC, Desterro JMP, Ferreira J, de Almeida SF. SETD2 is required for DNA double-strand break repair and activation of the p53-mediated checkpoint. eLife 2014; 3:e02482. [PMID: 24843002 PMCID: PMC4038841 DOI: 10.7554/elife.02482] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Histone modifications establish the chromatin states that coordinate the DNA damage response. In this study, we show that SETD2, the enzyme that trimethylates histone H3 lysine 36 (H3K36me3), is required for ATM activation upon DNA double-strand breaks (DSBs). Moreover, we find that SETD2 is necessary for homologous recombination repair of DSBs by promoting the formation of RAD51 presynaptic filaments. In agreement, SETD2-mutant clear cell renal cell carcinoma (ccRCC) cells displayed impaired DNA damage signaling. However, despite the persistence of DNA lesions, SETD2-deficient cells failed to activate p53, a master guardian of the genome rarely mutated in ccRCC and showed decreased cell survival after DNA damage. We propose that this novel SETD2-dependent role provides a chromatin bookmarking instrument that facilitates signaling and repair of DSBs. In ccRCC, loss of SETD2 may afford an alternative mechanism for the inactivation of the p53-mediated checkpoint without the need for additional genetic mutations in TP53. DOI:http://dx.doi.org/10.7554/eLife.02482.001 Normal wear and tear, exposure to chemicals, and ultraviolet light can all damage DNA, so cells rely on a range of sensors and mechanisms to detect and repair damaged DNA. Cells also package DNA molecules inside structures called histones to protect them against damage. Double-strand breaks—one of the most serious forms of DNA damage—are detected by an enzyme called ATM, and can be repaired in two ways. Bringing the broken strands back together is an obvious method, but it is also error prone. Using templates to generate new DNA to repair the damage is less prone to error, but it can only happen at certain times of the cell cycle. Some cancers are linked to the faulty repair of double-strand breaks. Moreover, a type of kidney cancer called clear cell renal carcinoma is linked to a lack of activity by a protein called p53, even in individuals who don't have mutations in the gene for this protein. However, many people with this type of cancer have mutations in the gene for a protein called SETD2. To investigate the links between SETD2 and DNA repair, Carvalho et al. compared cells with and without mutations in the gene for SETD2. It emerged that SETD2 must be present for DNA repair to take place: the SETD2 modifies the histones so that they can recruit the enzymes that repair the DNA via the template approach (which is relatively error free). SETD2 may be particularly important for repairing damage to genes without introducing errors. Carvalho et al. also show that mutations in SETD2 are sufficient to inactivate p53. The gene for this protein, which impedes the proliferation of cells with genomic aberrations, such as double-strand breaks, is mutated in most cancers. Overall the results help to illustrate how histone modifications and the DNA damage repair mechanisms and checkpoints work in concert to suppress cancer. DOI:http://dx.doi.org/10.7554/eLife.02482.002
Collapse
Affiliation(s)
- Sílvia Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Alexandra C Vítor
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Sreerama C Sridhara
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Filipa B Martins
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Ana C Raposo
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Joana M P Desterro
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - João Ferreira
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Sérgio F de Almeida
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
93
|
Balmer NV, Leist M. Epigenetics and transcriptomics to detect adverse drug effects in model systems of human development. Basic Clin Pharmacol Toxicol 2014; 115:59-68. [PMID: 24476462 DOI: 10.1111/bcpt.12203] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 01/16/2014] [Indexed: 01/01/2023]
Abstract
Prenatal exposure to environmental chemicals or drugs has been associated with functional or structural deficits and the development of diseases in later life. For example, developmental neurotoxicity (DNT) is triggered by lead, and this compound may predispose to neurodegenerative diseases in later life. The molecular memory for such late consequences of early exposure is not known, but epigenetic mechanisms (modification of the chromatin structure) could take this role. Examples and underlying mechanisms have been compiled here for the field of DNT. Moreover, we addressed the question as to what readout is suitable for addressing drug memory effects. We summarize how complex developmental processes can be modelled in vitro by using the differentiation of human stem cells. Although cellular models can never replicate the final human DNT phenotype, they can model the adverse effect that a chemical has on key biological processes essential for organ formation and function. Highly information-rich transcriptomics data may inform on these changes and form the bridge from in vitro models to human prediction. We compiled data showing that transcriptome analysis can indicate toxicity patterns of drugs. A crucial question to be answered in our systems is when and how transcriptome changes indicate adversity (as opposed to transient adaptive responses), and how drug-induced changes are perpetuated over time even after washout of the drug. We present evidence for the hypothesis that changes in the histone methylation pattern could represent the persistence detector of an early insult that is transformed to an adverse effect at later time-points in life.
Collapse
Affiliation(s)
- Nina V Balmer
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | | |
Collapse
|
94
|
Histone deacetylase inhibitors and epigenetic modifications as a novel strategy in renal cell carcinoma. Cancer J 2014; 19:333-40. [PMID: 23867515 DOI: 10.1097/ppo.0b013e3182a09e07] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Recent investigations of renal cell carcinoma (RCC) have revealed several epigenetic modifications, as well as alterations in the genes and enzymes that regulate these changes. Preclinical models have revealed that histone gene modifiers and epigenetic alterations may play a critical role in RCC tumorigenesis. Specific changes in DNA methylation and mutations of histone modifiers have been identified and may be associated with an aggressive phenotype. In addition, the potential of reversing the effects of these enzymes and hence reversing the cellular epigenetic landscape to a "normal phenotype" have led to an increasing interest in developing targeted chromatin remodeling agents. However, the translation of the understanding of these changes to the clinic for the treatment of RCC has posed significant challenges, partly due to tumor heterogeneity. This review describes the aberrant histone and DNA alterations recently reported in RCC and highlights the potential targeted chromatin remodeling therapies in the management of this disease.
Collapse
|
95
|
TFPI1 mediates resistance to doxorubicin in breast cancer cells by inducing a hypoxic-like response. PLoS One 2014; 9:e84611. [PMID: 24489651 PMCID: PMC3904823 DOI: 10.1371/journal.pone.0084611] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 11/16/2013] [Indexed: 11/19/2022] Open
Abstract
Thrombin and hypoxia are important players in breast cancer progression. Breast cancers often develop drug resistance, but mechanisms linking thrombin and hypoxia to drug resistance remain unresolved. Our studies using Doxorubicin (DOX) resistant MCF7 breast cancer cells reveals a mechanism linking DOX exposure with hypoxic induction of DOX resistance. Global expression changes between parental and DOX resistant MCF7 cells were examined. Westerns, Northerns and immunocytochemistry were used to validate drug resistance and differentially expressed genes. A cluster of genes involved in the anticoagulation pathway, with Tissue Factor Pathway Inhibitor 1 (TFPI1) the top hit, was identified. Plasmids overexpressing TFPI1 were utilized, and 1% O2 was used to test the effects of hypoxia on drug resistance. Lastly, microarray datasets from patients with drug resistant breast tumors were interrogated for TFPI1 expression levels. TFPI1 protein levels were found elevated in 3 additional DOX resistant cells lines, from humans and rats, indicating evolutionarily conservation of the effect. Elevated TFPI1 in DOX resistant cells was active, as thrombin protein levels were coincidentally low. We observed elevated HIF1α protein in DOX resistant cells, and in cells with forced expression of TFPI1, suggesting TFPI1 induces HIF1α. TFPI1 also induced c-MYC, c-SRC, and HDAC2 protein, as well as DOX resistance in parental cells. Growth of cells in 1% O2 induced elevated HIF1α, BCRP and MDR-1 protein, and these cells were resistant to DOX. Our in vitro results were consistent with in vivo patient datasets, as tumors harboring increased BCRP and MDR-1 expression also had increased TFPI1 expression. Our observations are clinically relevant indicating that DOX treatment induces an anticoagulation cascade, leading to inhibition of thrombin and the expression of HIF1α. This in turn activates a pathway leading to drug resistance.
Collapse
|
96
|
Cao L, Zhu L, Yang J, Su J, Ni J, Du Y, Liu D, Wang Y, Wang F, Jin J, Cai Y. Correlation of low expression of hMOF with clinicopathological features of colorectal carcinoma, gastric cancer and renal cell carcinoma. Int J Oncol 2014; 44:1207-14. [PMID: 24452485 DOI: 10.3892/ijo.2014.2266] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/07/2014] [Indexed: 11/06/2022] Open
Abstract
Human MOF (males absent on the first), as a histone acetyltransferase, is responsible for histone H4K16 acetylation in human cells. Recent studies have shown that the abnormal gene expression of hMOF is involved in certain primary cancers. Here, we first report the involvement of hMOF expression in clinically diagnosed primary colorectal carcinoma (CRC) and gastric cancer. Simultaneously, the correlation of hMOF expression and clinicopathological features in CRC, gastric cancer and renal cell carcinoma (RCC) was analyzed. The hMOF mRNA expression was assessed in 44 CRC, 16 gastric cancer and 47 RCC human tissue samples by quantitative PCR (qPCR). Statistical analysis of qPCR data revealed a significant reduction (>2-fold decrease) of hMOF gene expression in CRC, 57% (25/44), 94% (15/16) in gastric cancer and 74% (35/47) in RCC tissues of the patients. In patients with CRC, lymph node metastasis and tumor stage were associated with hMOF expression patterns. However, no significant association between hMOF expression and tumor types emerged (p>0.05). Interestingly, in patients with gastric cancer, although no statistically significant difference was found between adjacent (<2 cm away from the cancer tissue) and normal tissues (>5 cm away from the cancer tissue), >2-fold reduction of hMOF expression in adjacent tissues had already appeared in 35% of patients. In addition, low expression of hMOF was strongly correlated with tumor differentiation (p<0.05) and survival of patients with gastric cancer (p<0.001). While in patients with RCC, downregulation of hMOF was connected to ccRCC and tissues with T1 tumor status. Our results suggest that downregulation of hMOF may be common in cancer tissues, and may represent a novel biomarker for tumor diagnosis.
Collapse
Affiliation(s)
- Lingling Cao
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Lin Zhu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Jiaxing Yang
- Department of Gastrointestinal Surgery, The First Clinical Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jiaming Su
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Jinsong Ni
- Department of Pathology, The First Clinical Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yujun Du
- Department of Nephrology, The First Clinical Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Da Liu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yanfang Wang
- Department of Pathology, The First Clinical Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Fei Wang
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Jingji Jin
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yong Cai
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
97
|
|
98
|
Application of “Omics” Technologies to In Vitro Toxicology. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2014. [DOI: 10.1007/978-1-4939-0521-8_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
|
99
|
Lelièvre SA, Weaver CM. Global nutrition research: nutrition and breast cancer prevention as a model. Nutr Rev 2013; 71:742-52. [PMID: 24447199 PMCID: PMC3901298 DOI: 10.1111/nure.12075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The gene-environment interaction is paramount in light of the worldwide rise in incidence of chronic diseases, with cancers in the pole position. Diet is an environmental factor with potential to influence cancer onset by shaping the epigenome (i.e., the genome organization that controls the differential expression of genes). Yet, there is no consensus regarding how diet might help prevent breast cancer, the second most frequent malignancy globally. The complexity of breast cancers requires working on a global and multidisciplinary scale to further understand the relationship between breast cancer type, diet, and the epigenome. This article describes the International Breast Cancer & Nutrition collaboration as one such approach. A global endeavor brings the diversity necessary to pinpoint important diet-gene relationships. Being developed are models, detection and assessment tools, and funding and public policy frameworks necessary to advance primary prevention research for the benefit of all populations affected by breast cancer. This paradigm can be adapted to understanding diet-gene relationships for other chronic diseases.
Collapse
Affiliation(s)
- Sophie A. Lelièvre
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
- Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
- Women’s Global Health Institute, Purdue University, West Lafayette, IN, 47907, USA
- Department of Nutrition Science, Purdue University, West Lafayette, IN, 47907, USA
| | - Connie M. Weaver
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
- Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
- Women’s Global Health Institute, Purdue University, West Lafayette, IN, 47907, USA
- Department of Nutrition Science, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
100
|
Huffman K, Martinez ED. Pre-clinical studies of epigenetic therapies targeting histone modifiers in lung cancer. Front Oncol 2013; 3:235. [PMID: 24058902 PMCID: PMC3766830 DOI: 10.3389/fonc.2013.00235] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 08/27/2013] [Indexed: 12/19/2022] Open
Abstract
Treatment options for lung cancer patients have been generally limited to standard therapies or targeted interventions which involve a small number of known mutations. Although the targeted therapies are initially successful, they most often result in drug resistance, relapse, and mortality. We now know that the complexity of lung cancer comes not only from genomic changes, but also from aberrant epigenetic regulatory events. Epigenetic therapies have shown promise as single agents in the treatment of hematological malignancies but have yet to meet this expectation in solid tumors thus fostering researchers to pursue new approaches in the development and use of epigenetic interventions. Here, we review some recent pre-clinical findings involving the use of drugs targeting histone modifying enzymes both as single agents and as co-therapies against lung cancer. A greater understanding of the impact of these epigenetic compounds in lung cancer signaling is needed and further evaluation in vivo is warranted in several cases based on the pre-clinical activity of a subset of compounds discussed in this review, including drugs co-targeting HDACs and EGF receptor, targeting Brd4 and targeting Jumonji histone demethylases.
Collapse
Affiliation(s)
- Kenneth Huffman
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center , Dallas, TX , USA
| | | |
Collapse
|