1
|
Moos HK, Patel R, Flaherty SK, Loverde SM, Nikolova EN. H2A.Z facilitates Sox2-nucleosome interaction by promoting DNA and histone H3 tail mobility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641691. [PMID: 40093108 PMCID: PMC11908261 DOI: 10.1101/2025.03.06.641691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Epigenetic regulation of eukaryotic chromatin structure and function can be modulated by histone variants and post-translational modifications. The conserved variant H2A.Z has been functionally linked to pioneer factors Sox2 and Oct4 that open chromatin and initiate cell fate-specific expression programs. However, the molecular basis for their interaction remains unknown. Using biochemistry, nuclear magnetic resonance (NMR) spectroscopy and molecular dynamics (MD) simulations, we examine the role of H2A.Z nucleosome dynamics in pioneer factor binding. We find that H2A.Z facilitates Sox2 and Oct4 binding at distinct locations in 601 nucleosomes. We further link this to increased DNA accessibility and perturbed dynamics of the H3 N-terminal tail, which we show competes with Sox2 for DNA binding. Our simulations validate a coupling between H2A.Z-mediated DNA unwrapping and altered H3 N-tail conformations with fewer contacts to DNA and the H2A.Z C- terminal tail. This destabilizing effect of H2A.Z is DNA sequence dependent and enhanced with the less stable Lin28B nucleosome. Collectively, our findings suggest that H2A.Z promotes pioneer factor binding by increasing access to DNA and reducing competition with H3 tails. This could have broader implications for how epigenetic marks or oncogenic mutations tune pioneer factor engagement with chromatin and thus affect its structure and recognition.
Collapse
|
2
|
Liu W, Padhi A, Zhang X, Narendran J, Anastasio MA, Nain AS, Irudayaraj J. Dynamic Heterochromatin States in Anisotropic Nuclei of Cells on Aligned Nanofibers. ACS NANO 2022; 16:10754-10767. [PMID: 35803582 PMCID: PMC9332347 DOI: 10.1021/acsnano.2c02660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The cancer cell nucleus deforms as it invades the interstitial spaces in tissues and the tumor microenvironment. While alteration of the chromatin structure in a deformed nucleus is expected and documented, the chromatin structure in the nuclei of cells on aligned matrices has not been elucidated. In this work we elucidate the spatiotemporal organization of heterochromatin in the elongated nuclei of cells on aligned nanofibers with stimulated emission depletion nanoscopy and fluorescence correlation spectroscopy. We show that the anisotropy of nuclei is sufficient to drive H3K9me3-heterochromatin alterations, with enhanced H3K9me3 nanocluster compaction and aggregation states that otherwise are indistinguishable from diffraction-limited microscopy. We interrogated the higher-order heterochromatin structures within major chromatin compartments in anisotropic nuclei and discovered a wider spatial dispersion of nanodomain clusters in the nucleoplasm and condensed larger nanoclusters near the periphery and pericentromeric heterochromatin. Upon examining the spatiotemporal dynamics of heterochromatin in anisotropic nuclei, we observed reduced mobility of the constitutive heterochromatin mark H3K9me3 and the associated heterochromatin protein 1 (HP1α) at the nucleoplasm and periphery regions, correlating with increased viscosity and changes in gene expression. Since heterochromatin remodeling is crucial to genome integrity, our results reveal an unconventional H3K9me3 heterochromatin distribution, providing cues to an altered chromatin state due to perturbations of the nuclei in aligned fiber configurations.
Collapse
Affiliation(s)
- Wenjie Liu
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, 1102 Everitt Lab, 1406 W. Green Street, Urbana, Illinois 61801, United States
- Biomedical
Research Center, Mills Breast Cancer Institute, Cancer Center at Illinois,
Micro and Nanotechnology Laboratory, Beckman
Institute, Carl Woese Institute for Genomic Biology, Urbana, Illinois 61801, United States
| | - Abinash Padhi
- Department
of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Xiaohui Zhang
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, 1102 Everitt Lab, 1406 W. Green Street, Urbana, Illinois 61801, United States
| | - Jairaj Narendran
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, 1102 Everitt Lab, 1406 W. Green Street, Urbana, Illinois 61801, United States
| | - Mark A. Anastasio
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, 1102 Everitt Lab, 1406 W. Green Street, Urbana, Illinois 61801, United States
| | - Amrinder S. Nain
- Department
of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Joseph Irudayaraj
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, 1102 Everitt Lab, 1406 W. Green Street, Urbana, Illinois 61801, United States
- Biomedical
Research Center, Mills Breast Cancer Institute, Cancer Center at Illinois,
Micro and Nanotechnology Laboratory, Beckman
Institute, Carl Woese Institute for Genomic Biology, Urbana, Illinois 61801, United States
| |
Collapse
|
3
|
Fujita S, Motoda Y, Kigawa T, Tsuchiya K, Numata K. Peptide-Based Polyion Complex Vesicles That Deliver Enzymes into Intact Plants To Provide Antibiotic Resistance without Genetic Modification. Biomacromolecules 2020; 22:1080-1090. [PMID: 33316156 DOI: 10.1021/acs.biomac.0c01380] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Direct delivery of enzymes into intact plants using cell-penetrating peptides (CPPs) is an attractive approach for modifying plant functions without genetic modification. However, by conventional methods, it is difficult to maintain the enzyme activity for a long time because of proteolysis of the enzymes under physiological conditions. Here, we developed a novel enzyme delivery system using polyion complex vesicles (PICsomes) to protect the enzyme from proteases. We created PICsome-bearing reactive groups at the surface by mixing an anionic block copolymer, alkyne-TEG-P(Lys-COOH), and a cationic peptide, P(Lys). The PICsome encapsulated neomycin phosphotransferase II (NPTII), a kanamycin resistance enzyme, and protected NPTII from proteases in vitro. A CPP-modified PICsome delivered NPTII into the root hair cells of Arabidopsis thaliana seedlings and provided kanamycin resistance in the seedlings that lasted for 7 days. Thus, the PICsome-mediated enzyme delivery system is a promising method for imparting long-term transient traits to plants without genetic modification.
Collapse
Affiliation(s)
- Seiya Fujita
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Yoko Motoda
- Biomacromolecules Research Team, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Takanori Kigawa
- RIKEN Center for Biosystems Dynamics Research, Laboratory for Cellular Structural Biology, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Kousuke Tsuchiya
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Keiji Numata
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.,Biomacromolecules Research Team, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
4
|
Lehmann K, Felekyan S, Kühnemuth R, Dimura M, Tóth K, Seidel CAM, Langowski J. Dynamics of the nucleosomal histone H3 N-terminal tail revealed by high precision single-molecule FRET. Nucleic Acids Res 2020; 48:1551-1571. [PMID: 31956896 PMCID: PMC7026643 DOI: 10.1093/nar/gkz1186] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/29/2019] [Accepted: 12/10/2019] [Indexed: 02/06/2023] Open
Abstract
Chromatin compaction and gene accessibility are orchestrated by assembly and disassembly of nucleosomes. Although the disassembly process was widely studied, little is known about the structure and dynamics of the disordered histone tails, which play a pivotal role for nucleosome integrity. This is a gap filling experimental FRET study from the perspective of the histone H3 N-terminal tail (H3NtT) of reconstituted mononucleosomes. By systematic variation of the labeling positions we monitored the motions of the H3NtT relative to the dyad axis and linker DNA. Single-molecule FRET unveiled that H3NtTs do not diffuse freely but follow the DNA motions with multiple interaction modes with certain permitted dynamic transitions in the μs to ms time range. We also demonstrate that the H3NtT can allosterically sense charge-modifying mutations within the histone core (helix α3 of histone H2A (R81E/R88E)) resulting in increased dynamic transitions and lower rate constants. Those results complement our earlier model on the NaCl induced nucleosome disassembly as changes in H3NtT configurations coincide with two major steps: unwrapping of one linker DNA and weakening of the internal DNA - histone interactions on the other side. This emphasizes the contribution of the H3NtT to the fine-tuned equilibrium between overall nucleosome stability and DNA accessibility.
Collapse
Affiliation(s)
- Kathrin Lehmann
- Division Biophysics of Macromolecules, German Cancer Research Center, Heidelberg D-69120, Germany.,Lehrstuhl für Molekulare Physikalische Chemie, Heinrich-Heine-Universität, Düsseldorf D-40225, Germany
| | - Suren Felekyan
- Lehrstuhl für Molekulare Physikalische Chemie, Heinrich-Heine-Universität, Düsseldorf D-40225, Germany
| | - Ralf Kühnemuth
- Lehrstuhl für Molekulare Physikalische Chemie, Heinrich-Heine-Universität, Düsseldorf D-40225, Germany
| | - Mykola Dimura
- Lehrstuhl für Molekulare Physikalische Chemie, Heinrich-Heine-Universität, Düsseldorf D-40225, Germany
| | - Katalin Tóth
- Division Biophysics of Macromolecules, German Cancer Research Center, Heidelberg D-69120, Germany
| | - Claus A M Seidel
- Lehrstuhl für Molekulare Physikalische Chemie, Heinrich-Heine-Universität, Düsseldorf D-40225, Germany
| | - Jörg Langowski
- Division Biophysics of Macromolecules, German Cancer Research Center, Heidelberg D-69120, Germany
| |
Collapse
|
5
|
Liu W, Biton E, Pathania A, Matityahu A, Irudayaraj J, Onn I. Monomeric cohesin state revealed by live-cell single-molecule spectroscopy. EMBO Rep 2020; 21:e48211. [PMID: 31886609 PMCID: PMC7001500 DOI: 10.15252/embr.201948211] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/17/2022] Open
Abstract
The cohesin complex plays an important role in the maintenance of genome stability. Cohesin is composed of four core subunits and a set of regulatory subunits that interact with the core subunits. Less is known about cohesin dynamics in live cells and on the contribution of individual subunits to the overall complex. Understanding the tethering mechanism of cohesin is still a challenge, especially because the proposed mechanisms are still not conclusive. Models proposed to describe tethering depend on either the monomeric cohesin ring or a cohesin dimer. Here, we investigate the role of cohesin dynamics and stoichiometry in live yeast cells at single-molecule resolution. We explore the effect of regulatory subunit deletion on cohesin mobility and found that depletion of different regulatory subunits has opposing effects. Finally, we show that cohesin exists mostly as a canonical monomer throughout the cell cycle, and its monomeric form is independent of its regulatory factors. Our results demonstrate that single-molecule tools have the potential to provide new insights into the cohesin mechanism of action in live cells.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Bioengineering, Micro and Nanotechnology LaboratoryCancer Center at IllinoisUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Mills Breast Cancer InstituteCarle Foundation HospitalUrbanaILUSA
| | - Elisheva Biton
- The Azrieli Faculty of MedicineBar‐Ilan UniversitySafedIsrael
| | - Anjali Pathania
- The Azrieli Faculty of MedicineBar‐Ilan UniversitySafedIsrael
| | - Avi Matityahu
- The Azrieli Faculty of MedicineBar‐Ilan UniversitySafedIsrael
| | - Joseph Irudayaraj
- Department of Bioengineering, Micro and Nanotechnology LaboratoryCancer Center at IllinoisUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Mills Breast Cancer InstituteCarle Foundation HospitalUrbanaILUSA
| | - Itay Onn
- The Azrieli Faculty of MedicineBar‐Ilan UniversitySafedIsrael
| |
Collapse
|
6
|
Liu W, Irudayaraj J. Understanding the dynamics and structure of epigenetic states with single-molecule fluorescence microscopy. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019. [DOI: 10.1016/j.cobme.2019.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
7
|
Gutiérrez MP, MacAlpine HK, MacAlpine DM. Nascent chromatin occupancy profiling reveals locus- and factor-specific chromatin maturation dynamics behind the DNA replication fork. Genome Res 2019; 29:1123-1133. [PMID: 31217252 PMCID: PMC6633257 DOI: 10.1101/gr.243386.118] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 05/28/2019] [Indexed: 01/06/2023]
Abstract
Proper regulation and maintenance of the epigenome is necessary to preserve genome function. However, in every cell division, the epigenetic state is disassembled and then reassembled in the wake of the DNA replication fork. Chromatin restoration on nascent DNA is a complex and regulated process that includes nucleosome assembly and remodeling, deposition of histone variants, and the re-establishment of transcription factor binding. To study the genome-wide dynamics of chromatin restoration behind the DNA replication fork, we developed nascent chromatin occupancy profiles (NCOPs) to comprehensively profile nascent and mature chromatin at nucleotide resolution. Although nascent chromatin is inherently less organized than mature chromatin, we identified locus-specific differences in the kinetics of chromatin maturation that were predicted by the epigenetic landscape, including the histone variant H2AZ, which marked loci with rapid maturation kinetics. The chromatin maturation at origins of DNA replication was dependent on whether the origin underwent initiation or was passively replicated from distal-originating replication forks, suggesting distinct chromatin assembly mechanisms surrounding activated and disassembled prereplicative complexes. Finally, we identified sites that were only occupied transiently by DNA-binding factors following passage of the replication fork, which may provide a mechanism for perturbations of the DNA replication program to shape the regulatory landscape of the genome.
Collapse
Affiliation(s)
- Mónica P Gutiérrez
- University Program in Genetics and Genomics, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Heather K MacAlpine
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - David M MacAlpine
- University Program in Genetics and Genomics, Duke University Medical Center, Durham, North Carolina 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
8
|
Giaimo BD, Ferrante F, Herchenröther A, Hake SB, Borggrefe T. The histone variant H2A.Z in gene regulation. Epigenetics Chromatin 2019; 12:37. [PMID: 31200754 PMCID: PMC6570943 DOI: 10.1186/s13072-019-0274-9] [Citation(s) in RCA: 201] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 04/23/2019] [Indexed: 01/04/2023] Open
Abstract
The histone variant H2A.Z is involved in several processes such as transcriptional control, DNA repair, regulation of centromeric heterochromatin and, not surprisingly, is implicated in diseases such as cancer. Here, we review the recent developments on H2A.Z focusing on its role in transcriptional activation and repression. H2A.Z, as a replication-independent histone, has been studied in several model organisms and inducible mammalian model systems. Its loading machinery and several modifying enzymes have been recently identified, and some of the long-standing discrepancies in transcriptional activation and/or repression are about to be resolved. The buffering functions of H2A.Z, as supported by genome-wide localization and analyzed in several dynamic systems, are an excellent example of transcriptional control. Posttranslational modifications such as acetylation and ubiquitination of H2A.Z, as well as its specific binding partners, are in our view central players in the control of gene expression. Understanding the key-mechanisms in either turnover or stabilization of H2A.Z-containing nucleosomes as well as defining the H2A.Z interactome will pave the way for therapeutic applications in the future.
Collapse
Affiliation(s)
| | - Francesca Ferrante
- Institute of Biochemistry, University of Giessen, Friedrichstrasse 24, 35392, Giessen, Germany
| | - Andreas Herchenröther
- Institute for Genetics, University of Giessen, Heinrich-Buff-Ring 58-62, 35392, Giessen, Germany
| | - Sandra B Hake
- Institute for Genetics, University of Giessen, Heinrich-Buff-Ring 58-62, 35392, Giessen, Germany
| | - Tilman Borggrefe
- Institute of Biochemistry, University of Giessen, Friedrichstrasse 24, 35392, Giessen, Germany.
| |
Collapse
|
9
|
Damayanti NP, Buno K, Voytik Harbin SL, Irudayaraj JMK. Epigenetic Process Monitoring in Live Cultures with Peptide Biosensors. ACS Sens 2019; 4:562-565. [PMID: 30714727 DOI: 10.1021/acssensors.8b01134] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Acetyltransferase is a member of the transferase group responsible for transferring an acetyl group from acetyl-CoA to amino group of a histone lysine residue. Past efforts on histone acetylation monitoring involved biochemical analysis that do not provide spatiotemporal information in a dynamic format. We propose a novel approach to monitor acetyltransferase acetylation in live single cells using time correlated single photon counting fluorescence lifetime imaging (TCSPC-FLIM) with peptide biosensors. Utilizing 2D and 3D cultures we show that the peptide sensor has a specific response to acetyltransferase enzyme activity in a fluorescence lifetime dependent manner ( P < 0.001). Our FLIM biosensor concept enables real-time longitudinal measurement of acetylation activity with high spatial and temporal resolution in live single cells to monitor cell function or evaluate drug effects to treat cancer or neurological diseases.
Collapse
Affiliation(s)
- Nur P. Damayanti
- Department of Agricultural and Biological Engineering, College of Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Kevin Buno
- Weldon School of Biomedical Engineering, Collage of Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Sherry L. Voytik Harbin
- Weldon School of Biomedical Engineering, Collage of Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Joseph M. K. Irudayaraj
- Department of
Bioengineering, Cancer Center at Illinois, University of Illinois
at Urbana−Champaign, Biomedical Research Center, Carle Foundation
Hospital, Urbana, Illinois 61801, United States
| |
Collapse
|
10
|
Modulation of Gene Silencing by Cdc7p via H4 K16 Acetylation and Phosphorylation of Chromatin Assembly Factor CAF-1 in Saccharomyces cerevisiae. Genetics 2019; 211:1219-1237. [PMID: 30728156 DOI: 10.1534/genetics.118.301858] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/29/2019] [Indexed: 11/18/2022] Open
Abstract
CAF-1 is an evolutionarily conserved H3/H4 histone chaperone that plays a key role in replication-coupled chromatin assembly and is targeted to the replication fork via interactions with PCNA, which, if disrupted, leads to epigenetic defects. In Saccharomyces cerevisiae, when the silent mating-type locus HMR contains point mutations within the E silencer, Sir protein association and silencing is lost. However, mutation of CDC7, encoding an S-phase-specific kinase, or subunits of the H4 K16-specific acetyltransferase complex SAS-I, restore silencing to this crippled HMR, HMR a e** Here, we observed that loss of Cac1p, the largest subunit of CAF-1, also restores silencing at HMR a e**, and silencing in both cac1Δ and cdc7 mutants is suppressed by overexpression of SAS2 We demonstrate Cdc7p and Cac1p interact in vivo in S phase, but not in G1, consistent with observed cell cycle-dependent phosphorylation of Cac1p, and hypoacetylation of chromatin at H4 K16 in both cdc7 and cac1Δ mutants. Moreover, silencing at HMR a e** is restored in cells expressing cac1p mutants lacking Cdc7p phosphorylation sites. We also discovered that cac1Δ and cdc7-90 synthetically interact negatively in the presence of DNA damage, but that Cdc7p phosphorylation sites on Cac1p are not required for responses to DNA damage. Combined, our results support a model in which Cdc7p regulates replication-coupled histone modification via a CAC1-dependent mechanism involving H4 K16ac deposition, and thereby silencing, while CAF-1-dependent replication- and repair-coupled chromatin assembly per se are functional in the absence of phosphorylation of Cdc7p consensus sites on CAF-1.
Collapse
|
11
|
Liu W, Cui Y, Ren W, Irudayaraj J. Epigenetic biomarker screening by FLIM-FRET for combination therapy in ER+ breast cancer. Clin Epigenetics 2019; 11:16. [PMID: 30700309 PMCID: PMC6354376 DOI: 10.1186/s13148-019-0620-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 01/21/2019] [Indexed: 01/13/2023] Open
Abstract
Hormone-dependent gene expression involves dynamic and orchestrated regulation of epigenome leading to a cancerous state. Estrogen receptor (ER)-positive breast cancer rely on chromatin remodeling and association with epigenetic factors in inducing ER-dependent oncogenesis and thus cell over-proliferation. The mechanistic differences between epigenetic regulation and hormone signaling provide an avenue for combination therapy of ER-positive breast cancer. We hypothesized that epigenetic biomarkers within single nucleosome proximity of ER-dependent genes could serve as potential therapeutic targets. We described here a Fluorescence lifetime imaging-based Förster resonance energy transfer (FLIM-FRET) methodology for biomarker screening that could facilitate combination therapy based on our study. We screened 11 epigenetic-related markers which include oxidative forms of DNA methylation, histone modifications, and methyl-binding domain proteins. Among them, we identified H4K12acetylation (H4K12ac) and H3K27 acetylation (H3K27ac) as potential epigenetic therapeutic targets. When histone acetyltransferase inhibitor targeting H4K12ac and H3K27ac was combined with tamoxifen, an enhanced therapeutic outcome was observed against ER-positive breast cancer both in vitro and in vivo. Together, we demonstrate a single molecule approach as an effective screening tool for devising targeted epigenetic therapy.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA
- Present Address: Department of Bioengineering, Cancer Center at Illinois, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Champaign, IL, 61801, USA
| | - Yi Cui
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA
- Earth and Biological Science Directorate, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Wen Ren
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA
| | - Joseph Irudayaraj
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA.
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA.
- Present Address: Department of Bioengineering, Cancer Center at Illinois, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Champaign, IL, 61801, USA.
| |
Collapse
|
12
|
High precision FRET studies reveal reversible transitions in nucleosomes between microseconds and minutes. Nat Commun 2018; 9:4628. [PMID: 30401903 PMCID: PMC6219519 DOI: 10.1038/s41467-018-06758-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 09/17/2018] [Indexed: 11/08/2022] Open
Abstract
Nucleosomes play a dual role in compacting the genome and regulating the access to DNA. To unravel the underlying mechanism, we study fluorescently labeled mononucleosomes by multi-parameter FRET measurements and characterize their structural and dynamic heterogeneity upon NaCl-induced destabilization. Species-selective fluorescence lifetime analysis and dynamic photon distribution analysis reveal intermediates during nucleosome opening and lead to a coherent structural and kinetic model. In dynamic octasomes and hexasomes the interface between the H2A-H2B dimers and the (H3-H4)2 tetramer opens asymmetrically by an angle of ≈20° on a 50 and 15 µs time scale, respectively. This is followed by a slower stepwise release of the dimers coupled with DNA unwrapping. A mutation (H2A-R81A) at the interface between H2A and H3 facilitates initial opening, confirming the central role of the dimer:tetramer interface for nucleosome stability. Partially opened states such as those described here might serve as convenient nucleation sites for DNA-recognizing proteins.
Collapse
|
13
|
Weinhouse C, Truong L, Meyer JN, Allard P. Caenorhabditis elegans as an emerging model system in environmental epigenetics. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2018; 59:560-575. [PMID: 30091255 PMCID: PMC6113102 DOI: 10.1002/em.22203] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/13/2018] [Accepted: 04/19/2018] [Indexed: 05/19/2023]
Abstract
The roundworm Caenorhabitis elegans has been an established model organism for the study of genetics and developmental biology, including studies of transcriptional regulation, since the 1970s. This model organism has continued to be used as a classical model system as the field of transcriptional regulation has expanded to include scientific advances in epigenetics and chromatin biology. In the last several decades, C. elegans has emerged as a powerful model for environmental toxicology, particularly for the study of chemical genotoxicity. Here, we outline the utility and applicability of C. elegans as a powerful model organism for mechanistic studies of environmental influences on the epigenome. Our goal in this article is to inform the field of environmental epigenetics of the strengths and limitations of the well-established C. elegans model organism as an emerging model for medium-throughput, in vivo exploration of the role of exogenous chemical stimuli in transcriptional regulation, developmental epigenetic reprogramming, and epigenetic memory and inheritance. As the field of environmental epigenetics matures, and research begins to map mechanisms underlying observed associations, new toolkits and model systems, particularly manipulable, scalable in vivo systems that accurately model human transcriptional regulatory circuits, will provide an essential experimental bridge between in vitro biochemical experiments and mammalian model systems. Environ. Mol. Mutagen. 59:560-575, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Caren Weinhouse
- Duke Global Health Institute, Duke University, Durham, North Carolina
- Nicholas School of the Environment, Duke University, Durham, North Carolina
| | - Lisa Truong
- UCLA Human Genetics and Genomic Analysis Training Program, University of California, Los Angeles; Los Angeles, California
| | - Joel N. Meyer
- Duke Global Health Institute, Duke University, Durham, North Carolina
- Nicholas School of the Environment, Duke University, Durham, North Carolina
| | - Patrick Allard
- Institute for Society and Genetics, University of California at Los Angeles, Los Angeles, California
| |
Collapse
|
14
|
Damayanti NP, Buno K, Cui Y, Voytik-Harbin SL, Pili R, Freeman J, Irudayaraj JMK. Real-Time Multiplex Kinase Phosphorylation Sensors in Living Cells. ACS Sens 2017; 2:1225-1230. [PMID: 28838242 DOI: 10.1021/acssensors.7b00359] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Phosphorylation is an important post-translational modification implicated in cellular signaling and regulation. However, current methods to study protein phosphorylation by various kinases lack spatiotemporal resolution or the ability to simultaneously observe in real time the activity of multiple kinases in live cells. We present a peptide biosensor strategy with time correlated single photon counting-fluorescence lifetime imaging (TCSPC-FLIM) to interrogate the spatial and temporal dynamics of VEGFR-2 and AKT phosphorylation activity in real time in live 2D and 3D cell culture models at single cell resolution. By recording the increase in fluorescence lifetime due to a change in the solvatochromic environment of the sensor upon phosphorylation, we demonstrate that spatiotemporal maps of protein kinase activity can be obtained. Our results suggest that fluorescence lifetime imaging of peptide biosensors can be effectively and specifically used to monitor and quantify phosphorylation of multiple kinases in live cells.
Collapse
Affiliation(s)
- Nur P. Damayanti
- Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | | | | | | | - Roberto Pili
- Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Jennifer Freeman
- School of Health Sciences, West Lafayette, Indiana 47907, United States
| | | |
Collapse
|
15
|
Damayanti NP, Buno K, Narayanan N, Voytik Harbin SL, Deng M, Irudayaraj JMK. Monitoring focal adhesion kinase phosphorylation dynamics in live cells. Analyst 2017; 142:2713-2716. [PMID: 28589989 PMCID: PMC5531600 DOI: 10.1039/c7an00471k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Focal adhesion kinase (FAK) is a cytoplasmic non-receptor tyrosine kinase essential for a diverse set of cellular functions. Current methods for monitoring FAK activity in response to an extracellular stimulus lack spatiotemporal resolution and/or the ability to perform multiplex detection. Here we report on a novel approach to monitor the real-time kinase phosphorylation activity of FAK in live single cells by fluorescence lifetime imaging.
Collapse
Affiliation(s)
- Nur P Damayanti
- Bindley Bioscience Center, Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA. and Indiana University School of Medicine, 980 West Walnut St. R3 C312, Indianapolis, Indiana 46202, USA
| | - Kevin Buno
- Weldon School of Biomedical Engineering, Collage of Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Nagarajan Narayanan
- Bindley Bioscience Center, Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA.
| | - Sherry L Voytik Harbin
- Weldon School of Biomedical Engineering, Collage of Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Meng Deng
- Bindley Bioscience Center, Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA.
| | - Joseph M K Irudayaraj
- Bindley Bioscience Center, Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA.
| |
Collapse
|
16
|
Naz A, Cui Y, Collins CJ, Thompson DH, Irudayaraj J. PLGA-PEG nano-delivery system for epigenetic therapy. Biomed Pharmacother 2017; 90:586-597. [PMID: 28407579 DOI: 10.1016/j.biopha.2017.03.093] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 12/31/2022] Open
Abstract
Efficient delivery of cytidine analogues such as Azacitidine (AZA) into solid tumors constitutes a primary challenge in epigenetic therapies. We developed a di-block nano-vector based on poly(lactic-co-glycolic acid) (PLGA) and poly(ethylene glycol) (PEG) for stabilization of the conjugated AZA under physiological conditions. With equimolar drug content, our nano-conjugate could elicit a better anti-proliferative effect over free drug in breast cancer both in vitro and in vivo, through reactivation of p21 and BRCA1 to restrict cell proliferation. In addition, we applied single-molecule fluorescence tools to characterize the intracellular behavior of the AZA-PLGE-PEG nano-micelles at a finer spatiotemporal resolution. Our results suggest that the nano-micelles could effectively enrich in cancer cells and may not be limited by nucleoside transporters. Afterwards, the internalized nano-micelles exhibit pH-dependent release and resistance to active efflux. Altogether, our work describes a delivery strategy for DNA demethylating agents with nanoscale tunability, providing a cost-effective option for pharmaceutics.
Collapse
Affiliation(s)
- Asia Naz
- Bindley Bioscience Center and Purdue Center for Cancer Research, Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA; Department of Pharmaceutical Chemistry, University of Karachi, Karachi 75270, Pakistan
| | - Yi Cui
- Bindley Bioscience Center and Purdue Center for Cancer Research, Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | | | - David H Thompson
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Joseph Irudayaraj
- Bindley Bioscience Center and Purdue Center for Cancer Research, Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
17
|
Chowdhury B, Cho IH, Irudayaraj J. Technical advances in global DNA methylation analysis in human cancers. J Biol Eng 2017; 11:10. [PMID: 28261325 PMCID: PMC5331624 DOI: 10.1186/s13036-017-0052-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 02/10/2017] [Indexed: 02/07/2023] Open
Abstract
Prototypical abnormalities of genome-wide DNA methylation constitute the most widely investigated epigenetic mechanism in human cancers. Errors in the cellular machinery to faithfully replicate the global 5-methylcytosine (5mC) patterns, commonly observed during tumorigenesis, give rise to misregulated biological pathways beneficial to the rapidly propagating tumor mass but deleterious to the healthy tissues of the affected individual. A growing body of evidence suggests that the global DNA methylation levels could serve as utilitarian biomarkers in certain cancer types. Important breakthroughs in the recent years have uncovered further oxidized derivatives of 5mC - 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC), thereby expanding our understanding of the DNA methylation dynamics. While the biological roles of these epigenetic derivatives are being extensively characterized, this review presents a perspective on the opportunity of innovation in the global methylation analysis platforms. While multiple methods for global analysis of 5mC in clinical samples exist and have been reviewed elsewhere, two of the established methods - Liquid Chromatography coupled with mass spectrometry (LC-MS/MS) and Immunoquantification have successfully evolved to include the quantitation of 5hmC, 5fC and 5caC. Although the analytical performance of LC-MS/MS is superior, the simplicity afforded by the experimental procedure of immunoquantitation ensures it’s near ubiquity in clinical applications. Recent developments in spectroscopy, nanotechnology and sequencing also provide immense promise for future evaluations and are discussed briefly. Finally, we provide a perspective on the current scenario of global DNA methylation analysis tools and present suggestions to develop the next generation toolset.
Collapse
Affiliation(s)
- Basudev Chowdhury
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, 47907 IN USA
| | - Il-Hoon Cho
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam, 461-713 Republic of Korea
| | - Joseph Irudayaraj
- Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907 USA
| |
Collapse
|
18
|
Choudhury SR, Cui Y, Narayanan A, Gilley DP, Huda N, Lo CL, Zhou FC, Yernool D, Irudayaraj J. Optogenetic regulation of site-specific subtelomeric DNA methylation. Oncotarget 2016; 7:50380-50391. [PMID: 27391261 PMCID: PMC5226589 DOI: 10.18632/oncotarget.10394] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/18/2016] [Indexed: 01/24/2023] Open
Abstract
Telomere length homeostasis, critical for chromosomal integrity and genome stability, is controlled by intricate molecular regulatory machinery that includes epigenetic modifications. Here, we examine site-specific and spatiotemporal alteration of the subtelomeric methylation of CpG islands using optogenetic tools to understand the epigenetic regulatory mechanisms of telomere length maintenance. Human DNA methyltransferase3A (DNMT3A) were assembled selectively at chromosome ends by fusion to cryptochrome 2 protein (CRY2) and its interacting complement, the basic helix loop helix protein-1 (CIB1). CIB1 was fused to the telomere-associated protein telomere repeat binding factor-1 (TRF1), which localized the protein complex DNMT3A-CRY2 at telomeric regions upon excitation by blue-light monitored by single-molecule fluorescence analyses. Increased methylation was achieved selectively at subtelomeric CpG sites on the six examined chromosome ends specifically after blue-light activation, which resulted in progressive increase in telomere length over three generations of HeLa cell replications. The modular design of the fusion constructs presented here allows for the selective substitution of other chromatin modifying enzymes and for loci-specific targeting to regulate the epigenetic pathways at telomeres and other selected genomic regions of interest.
Collapse
Affiliation(s)
- Samrat Roy Choudhury
- Department of Agricultural & Biological Engineering, Bindley Bioscience Center, Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Yi Cui
- Department of Agricultural & Biological Engineering, Bindley Bioscience Center, Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Anoop Narayanan
- Bindley Laboratory of Structural Biology, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA
| | - David P. Gilley
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nazmul Huda
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chiao-Ling Lo
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Feng C. Zhou
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Institute of Neuroscience Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Dinesh Yernool
- Bindley Laboratory of Structural Biology, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA
| | - Joseph Irudayaraj
- Department of Agricultural & Biological Engineering, Bindley Bioscience Center, Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
19
|
Wang X, Cui Y, Irudayaraj J. Single-Cell Quantification of Cytosine Modifications by Hyperspectral Dark-Field Imaging. ACS NANO 2015; 9:11924-32. [PMID: 26505210 PMCID: PMC4766098 DOI: 10.1021/acsnano.5b04451] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Epigenetic modifications on DNA, especially on cytosine, play a critical role in regulating gene expression and genome stability. It is known that the levels of different cytosine derivatives are highly dynamic and are regulated by a variety of factors that act on the chromatin. Here we report an optical methodology based on hyperspectral dark-field imaging (HSDFI) using plasmonic nanoprobes to quantify the recently identified cytosine modifications on DNA in single cells. Gold (Au) and silver (Ag) nanoparticles (NPs) functionalized with specific antibodies were used as contrast-generating agents due to their strong local surface plasmon resonance (LSPR) properties. With this powerful platform we have revealed the spatial distribution and quantity of 5-carboxylcytosine (5caC) at the different stages in cell cycle and demonstrated that 5caC was a stably inherited epigenetic mark. We have also shown that the regional density of 5caC on a single chromosome can be mapped due to the spectral sensitivity of the nanoprobes in relation to the interparticle distance. Notably, HSDFI enables an efficient removal of the scattering noises from nonspecifically aggregated nanoprobes, to improve accuracy in the quantification of different cytosine modifications in single cells. Further, by separating the LSPR fingerprints of AuNPs and AgNPs, multiplex detection of two cytosine modifications was also performed. Our results demonstrate HSDFI as a versatile platform for spatial and spectroscopic characterization of plasmonic nanoprobe-labeled nuclear targets at the single-cell level for quantitative epigenetic screening.
Collapse
|
20
|
Abstract
The modENCODE (Model Organism Encyclopedia of DNA Elements) Consortium aimed to map functional elements-including transcripts, chromatin marks, regulatory factor binding sites, and origins of DNA replication-in the model organisms Drosophila melanogaster and Caenorhabditis elegans. During its five-year span, the consortium conducted more than 2,000 genome-wide assays in developmentally staged animals, dissected tissues, and homogeneous cell lines. Analysis of these data sets provided foundational insights into genome, epigenome, and transcriptome structure and the evolutionary turnover of regulatory pathways. These studies facilitated a comparative analysis with similar data types produced by the ENCODE Consortium for human cells. Genome organization differs drastically in these distant species, and yet quantitative relationships among chromatin state, transcription, and cotranscriptional RNA processing are deeply conserved. Of the many biological discoveries of the modENCODE Consortium, we highlight insights that emerged from integrative studies. We focus on operational and scientific lessons that may aid future projects of similar scale or aims in other, emerging model systems.
Collapse
Affiliation(s)
- James B Brown
- Department of Statistics, University of California, Berkeley, California 94720;
| | | |
Collapse
|
21
|
Abstract
Visualization of biological processes and pathologic conditions at the cellular and tissue levels largely relies on the use of fluorescence intensity signals from fluorophores or their bioconjugates. To overcome the concentration dependency of intensity measurements, evaluate subtle molecular interactions, and determine biochemical status of intracellular or extracellular microenvironments, fluorescence lifetime (FLT) imaging has emerged as a reliable imaging method complementary to intensity measurements. Driven by a wide variety of dyes exhibiting stable or environment-responsive FLTs, information multiplexing can be readily accomplished without the need for ratiometric spectral imaging. With knowledge of the fluorescent states of the molecules, it is entirely possible to predict the functional status of biomolecules or microevironment of cells. Whereas the use of FLT spectroscopy and microscopy in biological studies is now well-established, in vivo imaging of biological processes based on FLT imaging techniques is still evolving. This review summarizes recent advances in the application of the FLT of molecular probes for imaging cells and small animal models of human diseases. It also highlights some challenges that continue to limit the full realization of the potential of using FLT molecular probes to address diverse biological problems and outlines areas of potential high impact in the future.
Collapse
Affiliation(s)
- Pinaki Sarder
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, St. Louis, Missouri 63110
| | - Dolonchampa Maji
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, St. Louis, Missouri 63110
- Department of Biomedical Engineering, Washington University School of Medicine, 4525 Scott Avenue, St. Louis, Missouri 63110
| | - Samuel Achilefu
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, St. Louis, Missouri 63110
- Department of Biomedical Engineering, Washington University School of Medicine, 4525 Scott Avenue, St. Louis, Missouri 63110
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 4525 Scott Avenue, St. Louis, Missouri 63110
| |
Collapse
|
22
|
Cui Y, Irudayaraj J. Dissecting the behavior and function of MBD3 in DNA methylation homeostasis by single-molecule spectroscopy and microscopy. Nucleic Acids Res 2015; 43:3046-55. [PMID: 25753672 PMCID: PMC4381056 DOI: 10.1093/nar/gkv098] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 01/29/2015] [Indexed: 12/14/2022] Open
Abstract
The detailed mechanism for DNA methylation homeostasis relies on an intricate regulatory network with a possible contribution from methyl-CpG-binding domain protein 3 (MBD3). In this study we examine the single-molecule behavior of MBD3 and its functional implication in balancing the activity of DNA methyltransferases (DNMTs). Besides a localization tendency to DNA demethylating sites, MBD3 experiences a concurrent transcription with DNMTs in cell cycle. Fluorescence lifetime correlation spectroscopy (FLCS) and photon counting histogram (PCH) were applied to characterize the chromatin binding kinetics and stoichiometry of MBD3 in different cell phases. In the G1-phase, MBD3, in the context of the Mi-2/NuRD (nucleosome remodeling deacetylase) complex, could adopt a salt-dependent homodimeric association with its target epigenomic loci. Along with cell cycle progression, utilizing fluorescence lifetime imaging microscopy-based Förster resonance energy transfer (FLIM-FRET) we revealed that a proportion of MBD3 and MBD2 would co-localize with DNMT1 during DNA maintenance methylation, providing a proofreading and protective mechanism against a possible excessive methylation by DNMT1. In accordance with our hypothesis, insufficient MBD3 induced by small interfering RNA (siRNA) was found to result in a global DNA hypermethylation as well as increased methylation in the promoter CpG islands (CGIs) of a number of cell cycle related genes.
Collapse
Affiliation(s)
- Yi Cui
- Department of Agricultural and Biological Engineering, 225 S. University Street, Purdue University, West Lafayette, IN 47907, USA Bindley Bioscience Center, 1203 W. State Street, Purdue University, West Lafayette, IN 47907, USA
| | - Joseph Irudayaraj
- Department of Agricultural and Biological Engineering, 225 S. University Street, Purdue University, West Lafayette, IN 47907, USA Bindley Bioscience Center, 1203 W. State Street, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
23
|
Cui Y, Irudayaraj J. Inside single cells: quantitative analysis with advanced optics and nanomaterials. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2014; 7:387-407. [PMID: 25430077 DOI: 10.1002/wnan.1321] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/17/2014] [Accepted: 10/29/2014] [Indexed: 01/08/2023]
Abstract
Single-cell explorations offer a unique window to inspect molecules and events relevant to mechanisms and heterogeneity constituting the central dogma of biology. A large number of nucleic acids, proteins, metabolites, and small molecules are involved in determining and fine-tuning the state and function of a single cell at a given time point. Advanced optical platforms and nanotools provide tremendous opportunities to probe intracellular components with single-molecule accuracy, as well as promising tools to adjust single-cell activity. To obtain quantitative information (e.g., molecular quantity, kinetics, and stoichiometry) within an intact cell, achieving the observation with comparable spatiotemporal resolution is a challenge. For single-cell studies, both the method of detection and the biocompatibility are critical factors as they determine the feasibility, especially when considering live-cell analysis. Although a considerable proportion of single-cell methodologies depend on specialized expertise and expensive instruments, it is our expectation that the information content and implication will outweigh the costs given the impact on life science enabled by single-cell analysis.
Collapse
Affiliation(s)
- Yi Cui
- Department of Agricultural and Biological Engineering, Bindley Bioscience Center and Birck Nanotechnology Center, Purdue University, West Lafayette, IN, USA
| | | |
Collapse
|
24
|
Cui Y, Choudhury SR, Irudayaraj J. Quantitative real-time kinetics of optogenetic proteins CRY2 and CIB1/N using single-molecule tools. Anal Biochem 2014; 458:58-60. [PMID: 24780222 PMCID: PMC4341968 DOI: 10.1016/j.ab.2014.04.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/16/2014] [Accepted: 04/18/2014] [Indexed: 11/20/2022]
Abstract
In this work we evaluate the interaction of two optogenetic protein variants (CIB1, CIBN) with their complementary protein CRY2 by single-molecule tools in cell-free extracts. After validating the blue light induced co-localization of CRY2 and CIB1/N by Förster resonance energy transfer (FRET) in live cells, a fluorescence correlation spectroscopy (FCS) based method was developed to quantitatively determine the in vitro association of the extracted proteins. Our experiments suggest that CIB1, in comparison with CIBN, possesses a better coupling efficiency with CRY2 due to its intact protein structure and lower diffusion rate within 300s detection window.
Collapse
Affiliation(s)
- Yi Cui
- Department of Agricultural and Biological Engineering, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, United States
| | - Samrat Roy Choudhury
- Department of Agricultural and Biological Engineering, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, United States
| | - Joseph Irudayaraj
- Department of Agricultural and Biological Engineering, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
25
|
An RNA polymerase II-coupled function for histone H3K36 methylation in checkpoint activation and DSB repair. Nat Commun 2014; 5:3965. [PMID: 24910128 PMCID: PMC4052371 DOI: 10.1038/ncomms4965] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 04/25/2014] [Indexed: 12/20/2022] Open
Abstract
Histone modifications are major determinants of DNA double-strand break (DSB) response and repair. Here we elucidate a DSB repair function for transcription-coupled Set2 methylation at H3 lysine 36 (H3K36me). Cells devoid of Set2/H3K36me are hypersensitive to DNA-damaging agents and site-specific DSBs, fail to properly activate the DNA-damage checkpoint, and show genetic interactions with DSB-sensing and repair machinery. Set2/H3K36me3 is enriched at DSBs, and loss of Set2 results in altered chromatin architecture and inappropriate resection during G1 near break sites. Surprisingly, Set2 and RNA polymerase II are programmed for destruction after DSBs in a temporal manner – resulting in H3K36me3 to H3K36me2 transition that may be linked to DSB repair. Finally, we show a requirement of Set2 in DSB repair in transcription units – thus underscoring the importance of transcription-dependent H3K36me in DSB repair.
Collapse
|
26
|
Damayanti NP, Craig AP, Irudayaraj J. A hybrid FLIM-elastic net platform for label free profiling of breast cancer. Analyst 2014; 138:7127-34. [PMID: 24106733 DOI: 10.1039/c3an01097j] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report a label-free fluorescence lifetime profiling strategy to classify breast cancer cells, MCF10CA1h (malignant), MCF10A (nonmalignant), and MCF10AneoT (premalignant) in different stages of malignancy. Fluorescence Lifetime Imaging Microscopy (FLIM) was used to record the lifetime of autofluorescence of endogenous flavin in MCF10 cells in different stages of malignancy. Predominant differences in lifetimes ascertained by multi-exponential fitting curves can be attributed to the different forms of flavin protein; flavin mononucleotide (FMN), free flavin adenine dinucleotide (FAD), semiquinone, and bound FAD. A lifetime map of the metabolite was derived from the contribution of the lifetime of each metabolite by iterative reconvolution fitting of the Time Correlated Single Photon Counting (TCSPC) decay curves. Lifetime maps were constructed by mapping the average lifetime values pixel by pixel using MATLAB. The FLIM image (150 × 150 pixels) of each cell was extracted, resized and centered into 100 × 100 pixels using the nearest neighbor algorithm. Principal Component Analysis (PCA) in conjunction with Elastic net Analysis (EnA) was then used to classify the different stages of MCF10 cell lines based on average lifetime values. The EnA model provided an excellent classification of the cells at different stages of tumorigenesis yielding 100% accuracy.
Collapse
Affiliation(s)
- Nur P Damayanti
- Department of Agricultural and Biological Engineering and Bindley Bioscience Center, 225 S. University Street, West Lafayette, IN, USA
| | | | | |
Collapse
|
27
|
Vidi PA, Leary JF, Lelièvre SA. Building risk-on-a-chip models to improve breast cancer risk assessment and prevention. Integr Biol (Camb) 2014; 5:1110-8. [PMID: 23681255 DOI: 10.1039/c3ib40053k] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Preventive actions for chronic diseases hold the promise of improving lives and reducing healthcare costs. For several diseases, including breast cancer, multiple risk and protective factors have been identified by epidemiologists. The impact of most of these factors has yet to be fully understood at the organism, tissue, cellular and molecular levels. Importantly, combinations of external and internal risk and protective factors involve cooperativity thus, synergizing or antagonizing disease onset. Models are needed to mechanistically decipher cancer risks under defined cellular and microenvironmental conditions. Here, we briefly review breast cancer risk models based on 3D cell culture and propose to improve risk modeling with lab-on-a-chip approaches. We suggest epithelial tissue polarity, DNA repair and epigenetic profiles as endpoints in risk assessment models and discuss the development of 'risks-on-chips' integrating biosensors of these endpoints and of general tissue homeostasis. Risks-on-chips will help identify biomarkers of risk, serve as screening platforms for cancer preventive agents, and provide a better understanding of risk mechanisms, hence resulting in novel developments in disease prevention.
Collapse
Affiliation(s)
- Pierre-Alexandre Vidi
- Department of Basic Medical Sciences and Center for Cancer Research, Purdue University, 625 Harrison Street, Lynn Hall, West Lafayette, IN 47907-2026, USA.
| | | | | |
Collapse
|
28
|
Chen Y, Damayanti NP, Irudayaraj J, Dunn K, Zhou FC. Diversity of two forms of DNA methylation in the brain. Front Genet 2014; 5:46. [PMID: 24653733 PMCID: PMC3948076 DOI: 10.3389/fgene.2014.00046] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 02/12/2014] [Indexed: 11/13/2022] Open
Abstract
DNA methylation 5-methylcytosine (5mC) predicts a compacting chromatin inaccessible to transcription. The discovery of 5-hydroxymethylcytosine (5hmC), which is derived from 5mC, adds a new dimension to the mechanism and role of DNA methylation in epigenetics. Genomic evidence indicates that the 5hmC is located in the alternate regions to 5mC. However, the nature of 5hmC, as compared with classical 5mC remains unclear. Observing the mouse brain through embryonic development to the adult, first, we found that 5hmC is not merely an intermediate metabolite of demethylation, but is long lasting, chromatically distinct, and dynamically changing during neurodevelopment. Second, we found that 5hmC distinctly differs from 5mC in its chromatin affiliation during neural stem cell (NSC) development. Thirdly, we found both 5mC and 5hmC to be uniquely polarized and dynamic through the NSC development. 5mC was found to progressively polarize with MBD1 and MeCP2, and recruits H3K9me3 and H3K27me3; while 5hmC progressively co-localizes with MBD3 and recruits H3K4me2. Critical differential binding of 5mC with MBD1, and 5hmC with MBD3 was validated by Resonance Energy Transfer technique FLIM-FRET. This transition and polarization coincides with neuroprogenitor differentiation. Finally, at the time of synaptogenesis, 5mC gradually accumulates in the heterochromatin while 5hmC accumulates in the euchromatin, which is consistent with the co-localization of 5hmC with PolII, which mediates RNA transcription. Our data indicate that 5mC and 5hmC are diverse in their functional interactions with chromatin. This diversity is likely to contribute to the versatile epigenetic control of transcription mediating brain development and functional maintenance of adult brain.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine Indianapolis, IN, USA
| | - Nur P Damayanti
- Agricultural and Biological Engineering, Bindley Bioscience Center, Purdue University West Lafayette, IN, USA
| | - Joseph Irudayaraj
- Agricultural and Biological Engineering, Bindley Bioscience Center, Purdue University West Lafayette, IN, USA
| | - Kenneth Dunn
- Division of Nephology, Department of Medicine, Indiana University School of Medicine Indianapolis, IN, USA
| | - Feng C Zhou
- Department of Anatomy and Cell Biology, Indiana University School of Medicine Indianapolis, IN, USA ; Stark Neuroscience Research Institute, Indiana University School of Medicine Indianapolis, IN, USA
| |
Collapse
|
29
|
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: 0.9] [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
|
30
|
Cui Y, Cho IH, Chowdhury B, Irudayaraj J. Real-time dynamics of methyl-CpG-binding domain protein 3 and its role in DNA demethylation by fluorescence correlation spectroscopy. Epigenetics 2013; 8:1089-100. [PMID: 23974971 DOI: 10.4161/epi.25958] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
With unprecedented development in technology, epigenetics is recognized as a substantial and flexible regulatory pathway for phenotyping. Cytosine methylation and its subsequent oxidization have attracted significant attention due to their direct impact on gene regulation, in association with methyl-CpG-binding domain proteins (MBDs) and transcription related factors. In this study we record the dynamics of DNA demethylation using the recombinant MBD3-GFP protein in living cells under hypoxia and Decitabine treatment using Fluorescence Correlation Spectroscopy (FCS) by monitoring the diffusion dynamics of MBD3. Our study shows a DNA-replication-independent decrease of 5-methylcytosine (5mC)/5-hydroxymethylcytosine (5hmC) under hypoxia vs. a dependent decrease under Decitabine treatment. Further, we define a significantly faster diffusion of MBD3 in the nucleus as a precursory event for active demethylation rather than the Decitabine induced passive demethylation. By monitoring the diffusion of bound and unbound MBD3 in the nucleus we were able to identify and characterize hypoxia-sensitive cells from insensitive/tolerant cells, as well as the respective contribution to active demethylation in a time-dependent manner. Last, we quantitatively describe the concurrent decreasing trend in all of the three oxidized products of 5mC, which points to the potential involvement of ten-eleven-translocation proteins (TETs) in hypoxia induced active demethylation. Overall, for the first time we correlate the dynamic process of DNA demethylation with the biophysical properties of the corresponding DNA binding proteins in live single cells by single molecule spectroscopy.
Collapse
Affiliation(s)
- Yi Cui
- Biological Engineering and Bindley Bioscience Center, Purdue University; West Lafayette, IN USA
| | - Il-Hoon Cho
- Biological Engineering and Bindley Bioscience Center, Purdue University; West Lafayette, IN USA
| | - Basudev Chowdhury
- Biological Engineering and Bindley Bioscience Center, Purdue University; West Lafayette, IN USA
| | - Joseph Irudayaraj
- Biological Engineering and Bindley Bioscience Center, Purdue University; West Lafayette, IN USA
| |
Collapse
|
31
|
Banerjee A, Bhatia D, Saminathan A, Chakraborty S, Kar S, Krishnan Y. Controlled release of encapsulated cargo from a DNA icosahedron using a chemical trigger. Angew Chem Int Ed Engl 2013; 52:6854-7. [PMID: 23716499 DOI: 10.1002/anie.201302759] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Anusuya Banerjee
- Biochemistry and Biophysics, National Centre for Biological Sciences, UAS-GKVK, Bellary Road, Bangalore, India
| | | | | | | | | | | |
Collapse
|
32
|
Banerjee A, Bhatia D, Saminathan A, Chakraborty S, Kar S, Krishnan Y. Controlled Release of Encapsulated Cargo from a DNA Icosahedron using a Chemical Trigger. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201302759] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
33
|
Damayanti NP, Parker LL, Irudayaraj JMK. Fluorescence lifetime imaging of biosensor peptide phosphorylation in single live cells. Angew Chem Int Ed Engl 2013; 52:3931-4. [PMID: 23450802 DOI: 10.1002/anie.201209303] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 01/12/2013] [Indexed: 12/12/2022]
Affiliation(s)
- Nur P Damayanti
- Department of Agricultural and Biological Engineering, Bindley Bioscience Center, Purdue Center for Cancer Research, Purdue University, 225 S. University Street, West Lafayette, IN 47907, USA
| | | | | |
Collapse
|
34
|
Damayanti NP, Parker LL, Irudayaraj JMK. Fluorescence Lifetime Imaging of Biosensor Peptide Phosphorylation in Single Live Cells. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201209303] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|