1
|
Hamali B, Amine AAA, Al-Sady B. Regulation of the heterochromatin spreading reaction by trans-acting factors. Open Biol 2023; 13:230271. [PMID: 37935357 PMCID: PMC10645111 DOI: 10.1098/rsob.230271] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/03/2023] [Indexed: 11/09/2023] Open
Abstract
Heterochromatin is a gene-repressive protein-nucleic acid ultrastructure that is initially nucleated by DNA sequences. However, following nucleation, heterochromatin can then propagate along the chromatin template in a sequence-independent manner in a reaction termed spreading. At the heart of this process are enzymes that deposit chemical information on chromatin, which attracts the factors that execute chromatin compaction and transcriptional or co/post-transcriptional gene silencing. Given that these enzymes deposit guiding chemical information on chromatin they are commonly termed 'writers'. While the processes of nucleation and central actions of writers have been extensively studied and reviewed, less is understood about how the spreading process is regulated. We discuss how the chromatin substrate is prepared for heterochromatic spreading, and how trans-acting factors beyond writer enzymes regulate it. We examine mechanisms by which trans-acting factors in Suv39, PRC2, SETDB1 and SIR writer systems regulate spreading of the respective heterochromatic marks across chromatin. While these systems are in some cases evolutionarily and mechanistically quite distant, common mechanisms emerge which these trans-acting factors exploit to tune the spreading reaction.
Collapse
Affiliation(s)
- Bulut Hamali
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
- The G. W. Hooper Foundation, San Francisco, CA 94143, USA
- College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Ahmed A A Amine
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
- The G. W. Hooper Foundation, San Francisco, CA 94143, USA
| | - Bassem Al-Sady
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
- The G. W. Hooper Foundation, San Francisco, CA 94143, USA
| |
Collapse
|
2
|
Unconventional metabolites in chromatin regulation. Biosci Rep 2022; 42:230604. [PMID: 34988581 PMCID: PMC8777195 DOI: 10.1042/bsr20211558] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/04/2022] [Accepted: 01/04/2022] [Indexed: 11/17/2022] Open
Abstract
Chromatin, the complex of DNA and histone proteins, serves as a main integrator of cellular signals. Increasing evidence links cellular functional to chromatin state. Indeed, different metabolites are emerging as modulators of chromatin function and structure. Alterations in chromatin state are decisive for regulating all aspects of genome function and ultimately have the potential to produce phenotypic changes. Several metabolites such as acetyl-CoA, S-adenosylmethionine (SAM) or adenosine triphosphate (ATP) have now been well characterized as main substrates or cofactors of chromatin-modifying enzymes. However, there are other metabolites that can directly interact with chromatin influencing its state or that modulate the properties of chromatin regulatory factors. Also, there is a growing list of atypical enzymatic and nonenzymatic chromatin modifications that originate from different cellular pathways that have not been in the limelight of chromatin research. Here, we summarize different properties and functions of uncommon regulatory molecules originating from intermediate metabolism of lipids, carbohydrates and amino acids. Based on the various modes of action on chromatin and the plethora of putative, so far not described chromatin-regulating metabolites, we propose that there are more links between cellular functional state and chromatin regulation to be discovered. We hypothesize that these connections could provide interesting starting points for interfering with cellular epigenetic states at a molecular level.
Collapse
|
3
|
The Capability of O-Acetyl-ADP-Ribose, an Epigenetic Metabolic Small Molecule, on Promoting the Further Spreading of Sir3 along the Telomeric Chromatin. Genes (Basel) 2019; 10:genes10080577. [PMID: 31366171 PMCID: PMC6723988 DOI: 10.3390/genes10080577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/23/2019] [Accepted: 07/27/2019] [Indexed: 11/17/2022] Open
Abstract
O-acetyl-ADP-ribose (AAR) is a metabolic small molecule relevant in epigenetics that is generated by NAD-dependent histone deacetylases, such as Sir2. The formation of silent heterochromatin in yeast requires histone deacetylation by Sir2, structural rearrangement of SIR complexes, spreading of SIR complexes along the chromatin, and additional maturation processing. AAR affects the interactions of the SIR-nucleosome in vitro and enhances the chromatin epigenetic silencing effect in vivo. In this study, using isothermal titration calorimetry (ITC) and dot blotting methods, we showed the direct interaction of AAR with Sir3. Furthermore, through chromatin immunoprecipitation (ChIP)-on-chip and chromatin affinity purification (ChAP)-on chip assays, we discovered that AAR is capable of increasing the extended spreading of Sir3 along telomeres, but not Sir2. In addition, the findings of a quantitative real-time polymerase chain reaction (qRT-PCR) and examinations of an in vitro assembly system of SIR-nucleosome heterochromatin filament were consistent with these results. This study provides evidence indicating another important effect of AAR in vivo. AAR may play a specific modulating role in the formation of silent SIR-nucleosome heterochromatin in yeast.
Collapse
|
4
|
Wang SH, Lee SP, Tung SY, Tsai SP, Tsai HC, Shen HH, Hong JY, Su KC, Chen FJ, Liu BH, Wu YY, Hsiao SP, Tsai MS, Liou GG. Stabilization of Sir3 interactions by an epigenetic metabolic small molecule, O-acetyl-ADP-ribose, on yeast SIR-nucleosome silent heterochromatin. Arch Biochem Biophys 2019; 671:167-174. [PMID: 31295433 DOI: 10.1016/j.abb.2019.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/04/2019] [Accepted: 07/05/2019] [Indexed: 11/24/2022]
Abstract
In Saccharomyces cerevisiae, Sir proteins mediate heterochromatin epigenetic gene silencing. The assembly of silent heterochromatin requires histone deacetylation by Sir2, conformational change of SIR complexes, and followed by spreading of SIR complexes along the chromatin fiber to form extended silent heterochromatin domains. Sir2 couples histone deacetylation and NAD hydrolysis to generate an epigenetic metabolic small molecule, O-acetyl-ADP-ribose (AAR). Here, we demonstrate that AAR physically associates with Sir3 and that polySir3-AAR formation has a specific and essential role in the assembly of silent SIR-nucleosome pre-heterochromatin filaments. Furthermore, we show that AAR is capable of stabilizing binding of the Sir3 BAH domain to the Sir3 carboxyl-terminal region. Our data suggests that for the assembly of SIR-nucleosome pre-heterochromatin filament, the structural rearrangement of SIR-nucleosome is important and result in creating more stable interactions of Sir3, such as the inter-molecule Sir3-Sir3 interaction, and the Sir3-nucleosome interaction within the filaments. In conclusion, our results reveal the importance of AAR, indicating that it not only affects the conformational rearrangement of SIR complexes but also might function as a critical fine-tuning modulatory component of yeast silent SIR-nucleosome pre-heterochromatin by stabilizing the intermolecular interaction between Sir3 N- and C-terminal regions.
Collapse
Affiliation(s)
- Sue-Hong Wang
- Department of Biomedical Sciences, Chung Shan Medical University & Department of Medical Research, Chung Shan Medical University Hospital, Taichung, 402, Taiwan, ROC
| | - Sue-Ping Lee
- Institute of Molecular Biology, Academia Sinica, Taipei, 115, Taiwan, ROC
| | - Shu-Yun Tung
- Institute of Molecular Biology, Academia Sinica, Taipei, 115, Taiwan, ROC
| | - Shu-Ping Tsai
- Institute of Molecular Biology, Academia Sinica, Taipei, 115, Taiwan, ROC
| | - Hsieh-Chin Tsai
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, 350, Taiwan, ROC
| | - Hsiao-Hsuian Shen
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, 350, Taiwan, ROC
| | - Jia-Yang Hong
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, 350, Taiwan, ROC
| | - Kuan-Chung Su
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, 350, Taiwan, ROC
| | - Feng-Jung Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, 350, Taiwan, ROC
| | - Bang-Hung Liu
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, 350, Taiwan, ROC
| | - Yu-Yi Wu
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, 350, Taiwan, ROC
| | - Sheng-Pin Hsiao
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, 350, Taiwan, ROC
| | - Ming-Shiun Tsai
- Department of Food Science and Biotechnology, Da-Yeh University, Changhua, 515, Taiwan, ROC
| | - Gunn-Guang Liou
- Institute of Molecular Biology, Academia Sinica, Taipei, 115, Taiwan, ROC; Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, 350, Taiwan, ROC; Department of Food Science and Biotechnology, Da-Yeh University, Changhua, 515, Taiwan, ROC; Institute of Biological Chemistry, Academia Sinica, Taipei, 115, Taiwan, ROC; Guang EM Laboratory, New Taipei, 242, Taiwan, ROC.
| |
Collapse
|
5
|
Wang SH, Tung SY, Su KC, Shen HH, Hong JY, Tsai MS, Liou GG. Enhancer role of a native metabolite, O-acetyl-ADP-ribose, on the Saccharomyces cerevisiae chromatin epigenetic gene silencing. Genes Cells 2019; 24:449-457. [PMID: 30974043 DOI: 10.1111/gtc.12685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 03/26/2019] [Accepted: 04/05/2019] [Indexed: 11/30/2022]
Abstract
To study the epigenetic gene silencing, yeast is an excellent model organism. Sir proteins are required for the formation of silent heterochromatin. Sir2 couples histone deacetylation and NAD hydrolysis to generate an endogenous epigenetic metabolic small molecule, O-acetyl-ADP-ribose (AAR). AAR is involved in the conformational change of SIR complexes, modulates the formation of SIR-nucleosome preheterochromatin and contributes to the spreading of SIR complexes along the chromatin fiber to form extended silent heterochromatin regions. Here, we show that AAR is capable of enhancing the chromatin silencing effect under either an extra exogenous AAR or a defect AAR metabolic enzyme situation, but decreasing the chromatin silencing effect under a defect AAR synthetic enzyme state. Our results provide an evidence of biological function importance of AAR. It is indicated that AAR does not only function in vitro but also play a role in vivo to increase the effect of heterochromatin epigenetic gene silencing. However, further investigations of AAR are warranted to expand our knowledge of epigenetics and associated small molecules.
Collapse
Affiliation(s)
- Sue-Hong Wang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Shu-Yun Tung
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Kuan-Chung Su
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Hsiao-Hsuian Shen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Jia-Yang Hong
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ming-Shiun Tsai
- Department of Food Science and Biotechnology, Da-Yeh University, Changhua, Taiwan
| | - Gunn-Guang Liou
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.,Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan.,Department of Food Science and Biotechnology, Da-Yeh University, Changhua, Taiwan.,Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.,Guang EM Laboratory, New Taipei, Taiwan
| |
Collapse
|
6
|
Tung SY, Wang SH, Lee SP, Tsai SP, Shen HH, Chen FJ, Wu YY, Hsiao SP, Liou GG. Modulations of SIR-nucleosome interactions of reconstructed yeast silent pre-heterochromatin by O-acetyl-ADP-ribose and magnesium. Mol Biol Cell 2016; 28:381-386. [PMID: 27932495 PMCID: PMC5341722 DOI: 10.1091/mbc.e16-06-0359] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 11/22/2016] [Accepted: 11/29/2016] [Indexed: 12/25/2022] Open
Abstract
In vitro–assembled filaments are confirmed as SIR-nucleosome pre-heterochromatin, and AAR acts as a modulator for their formation. Not only is magnesium present in the environmental buffer, but it also is chelated by the SIR-nucleosome pre-heterochromatin to promote its condensation. Yeast silent heterochromatin provides an excellent model with which to study epigenetic inheritance. Previously we developed an in vitro assembly system to demonstrate the formation of filament structures with requirements that mirror yeast epigenetic gene silencing in vivo. However, the properties of these filaments were not investigated in detail. Here we show that the assembly system requires Sir2, Sir3, Sir4, nucleosomes, and O-acetyl-ADP-ribose. We also demonstrate that all Sir proteins and nucleosomes are components of these filaments to prove that they are SIR-nucleosome filaments. Furthermore, we show that the individual localization patterns of Sir proteins on the SIR-nucleosome filament reflect those patterns on telomeres in vivo. In addition, we reveal that magnesium exists in the SIR-nucleosome filament, with a role similar to that for chromatin condensation. These results suggest that a small number of proteins and molecules are sufficient to mediate the formation of a minimal yeast silent pre-heterochromatin in vitro.
Collapse
Affiliation(s)
- Shu-Yun Tung
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Sue-Hong Wang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung 402, Taiwan
| | - Sue-Ping Lee
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Shu-Ping Tsai
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Hsiao-Hsuian Shen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 350, Taiwan
| | - Feng-Jung Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 350, Taiwan
| | - Yu-Yi Wu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 350, Taiwan
| | - Sheng-Pin Hsiao
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 350, Taiwan
| | - Gunn-Guang Liou
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan .,Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 350, Taiwan.,Guang EM Laboratory, New Taipei 242, Taiwan
| |
Collapse
|
7
|
Erwin GS, Grieshop MP, Bhimsaria D, Eguchi A, Rodríguez-Martínez JA, Ansari AZ. Genome-wide Mapping of Drug-DNA Interactions in Cells with COSMIC (Crosslinking of Small Molecules to Isolate Chromatin). J Vis Exp 2016:e53510. [PMID: 26863565 DOI: 10.3791/53510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The genome is the target of some of the most effective chemotherapeutics, but most of these drugs lack DNA sequence specificity, which leads to dose-limiting toxicity and many adverse side effects. Targeting the genome with sequence-specific small molecules may enable molecules with increased therapeutic index and fewer off-target effects. N-methylpyrrole/N-methylimidazole polyamides are molecules that can be rationally designed to target specific DNA sequences with exquisite precision. And unlike most natural transcription factors, polyamides can bind to methylated and chromatinized DNA without a loss in affinity. The sequence specificity of polyamides has been extensively studied in vitro with cognate site identification (CSI) and with traditional biochemical and biophysical approaches, but the study of polyamide binding to genomic targets in cells remains elusive. Here we report a method, the crosslinking of small molecules to isolate chromatin (COSMIC), that identifies polyamide binding sites across the genome. COSMIC is similar to chromatin immunoprecipitation (ChIP), but differs in two important ways: (1) a photocrosslinker is employed to enable selective, temporally-controlled capture of polyamide binding events, and (2) the biotin affinity handle is used to purify polyamide-DNA conjugates under semi-denaturing conditions to decrease DNA that is non-covalently bound. COSMIC is a general strategy that can be used to reveal the genome-wide binding events of polyamides and other genome-targeting chemotherapeutic agents.
Collapse
Affiliation(s)
- Graham S Erwin
- Department of Biochemistry, University of Wisconsin-Madison
| | | | - Devesh Bhimsaria
- Department of Biochemistry, University of Wisconsin-Madison; Department of Electrical and Computer Engineering, University of Wisconsin-Madison
| | - Asuka Eguchi
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison
| | | | - Aseem Z Ansari
- Department of Biochemistry, University of Wisconsin-Madison; The Genome Center, University of Wisconsin-Madison;
| |
Collapse
|
8
|
Anders L, Guenther MG, Qi J, Fan ZP, Marineau JJ, Rahl PB, Lovén J, Sigova AA, Smith WB, Lee TI, Bradner JE, Young RA. Genome-wide localization of small molecules. Nat Biotechnol 2013; 32:92-6. [PMID: 24336317 PMCID: PMC4189815 DOI: 10.1038/nbt.2776] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/20/2013] [Indexed: 12/28/2022]
Abstract
A vast number of small-molecule ligands, including therapeutic drugs under development and in clinical use, elicit their effects by binding specific proteins associated with the genome. An ability to map the direct interactions of a chemical entity with chromatin genome-wide could provide important insights into chemical perturbation of cellular function. Here we describe a method that couples ligand-affinity capture and massively parallel DNA sequencing (Chem-seq) to identify the sites bound by small chemical molecules throughout the human genome. We show how Chem-seq can be combined with ChIP-seq to gain unique insights into the interaction of drugs with their target proteins throughout the genome of tumor cells. These methods will be broadly useful to enhance understanding of therapeutic action and to characterize the specificity of chemical entities that interact with DNA or genome-associated proteins.
Collapse
Affiliation(s)
- Lars Anders
- 1] Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA. [2]
| | - Matthew G Guenther
- 1] Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA. [2]
| | - Jun Qi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Massachusetts, USA
| | - Zi Peng Fan
- 1] Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA. [2] Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jason J Marineau
- Department of Medical Oncology, Dana-Farber Cancer Institute, Massachusetts, USA
| | - Peter B Rahl
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | - Jakob Lovén
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | - Alla A Sigova
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | - William B Smith
- Department of Medical Oncology, Dana-Farber Cancer Institute, Massachusetts, USA
| | - Tong Ihn Lee
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | - James E Bradner
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Massachusetts, USA. [2] Department of Medicine, Harvard Medical School, Massachusetts, USA
| | - Richard A Young
- 1] Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA. [2] Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
9
|
Kueng S, Oppikofer M, Gasser SM. SIR proteins and the assembly of silent chromatin in budding yeast. Annu Rev Genet 2013; 47:275-306. [PMID: 24016189 DOI: 10.1146/annurev-genet-021313-173730] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Saccharomyces cerevisiae provides a well-studied model system for heritable silent chromatin in which a histone-binding protein complex [the SIR (silent information regulator) complex] represses gene transcription in a sequence-independent manner by spreading along nucleosomes, much like heterochromatin in higher eukaryotes. Recent advances in the biochemistry and structural biology of the SIR-chromatin system bring us much closer to a molecular understanding of yeast silent chromatin. Simultaneously, genome-wide approaches have shed light on the biological importance of this form of epigenetic repression. Here, we integrate genetic, structural, and cell biological data into an updated overview of yeast silent chromatin assembly.
Collapse
Affiliation(s)
- Stephanie Kueng
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | | | | |
Collapse
|
10
|
Oppikofer M, Kueng S, Gasser SM. SIR–nucleosome interactions: Structure–function relationships in yeast silent chromatin. Gene 2013; 527:10-25. [DOI: 10.1016/j.gene.2013.05.088] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 05/27/2013] [Accepted: 05/30/2013] [Indexed: 01/09/2023]
|
11
|
Tung SY, Lee KW, Hong JY, Lee SP, Shen HH, Liou GG. Changes in the genome-wide localization pattern of Sir3 in Saccharomyces cerevisiae during different growth stages. Comput Struct Biotechnol J 2013; 7:e201304001. [PMID: 24688731 PMCID: PMC3962127 DOI: 10.5936/csbj.201304001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 05/14/2013] [Accepted: 05/19/2013] [Indexed: 12/03/2022] Open
Abstract
In budding yeast, the Sir2, Sir3 and Sir4 proteins form SIR complexes, required for the assembly of silent heterochromatin domains, and which mediate transcription silencing at the telomeres as well as at silent mating type loci. In this study, under fluorescence microscopy, we found most Sir3-GFP expressions in the logarithmic phase cells appeared as multiple punctations as expected. However, some differences in the distribution of fluorescent signals were detected in the diauxic~early stationary phase cells. To clarify these, we then used ChIP on chip assays to investigate the genome-wide localization of Sir3. In general, Sir3 binds to all 32 telomere proximal regions, the silent mating type loci and also binds to the rDNA region. However, the genome-wide localization patterns of Sir3 are different between these two distinct growth phases. We also confirmed that Sir3 binds to a recently identified secondary binding site, PAU genes, and further identified 349 Sir3-associated cluster regions. These results provide additional support in roles for Sir3 in the modulation of gene expression during physical conditions such as diauxic~early stationary phase growing. Moreover, they imply that Sir3 may be not only involved in the formation of conventional silent heterochromatin, but also able to associate with some other chromatin regions involved in epigenetic regulation.
Collapse
Affiliation(s)
- Shu-Yun Tung
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, ROC ; These authors contributed equally to this work
| | - Kuan-Wei Lee
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 35053, Taiwan, ROC ; These authors contributed equally to this work
| | - Jia-Yang Hong
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 35053, Taiwan, ROC ; These authors contributed equally to this work
| | - Sue-Ping Lee
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, ROC ; These authors contributed equally to this work
| | - Hsiao-Hsuian Shen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
| | - Gunn-Guang Liou
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 35053, Taiwan, ROC ; Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan, ROC
| |
Collapse
|