1
|
Jonas F, Navon Y, Barkai N. Intrinsically disordered regions as facilitators of the transcription factor target search. Nat Rev Genet 2025; 26:424-435. [PMID: 39984675 DOI: 10.1038/s41576-025-00816-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2025] [Indexed: 02/23/2025]
Abstract
Transcription factors (TFs) contribute to organismal development and function by regulating gene expression. Despite decades of research, the factors determining the specificity and speed at which eukaryotic TFs detect their target binding sites remain poorly understood. Recent studies have pointed to intrinsically disordered regions (IDRs) within TFs as key regulators of the process by which TFs find their target sites on DNA (the TF target search). However, IDRs are challenging to study because they can confer specificity despite low sequence complexity and can be functionally conserved despite rapid sequence divergence. Nevertheless, emerging computational and experimental approaches are beginning to elucidate the sequence-function relationship within the IDRs of TFs. Additional insights are informing potential mechanisms underlying the IDR-directed search for the DNA targets of TFs, including incorporation into biomolecular condensates, facilitating TF co-localization, and the hypothesis that IDRs recognize and directly interact with specific genomic regions.
Collapse
Affiliation(s)
- Felix Jonas
- School of Science, Constructor University, Bremen, Germany.
| | - Yoav Navon
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Naama Barkai
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
2
|
Lu Y, Wang FY, Levine MS, Shi HR, Wang Y, Xiong X, Yang LM, Shi YQ, Zou T, Sessler JL, Liang H, Huang KB. Oxoisoaporphine Alkaloid Iridium(III) Derivative: An Immunogenic Cell Death Inducer That Engages the Autophagy-Dependent Regulator Cathepsin D. J Am Chem Soc 2025; 147:15216-15228. [PMID: 40279467 DOI: 10.1021/jacs.5c00255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
Autophagy has been recognized as one of the pathways for eliciting immunogenic cell death (ICD). However, the specific molecular target responsible for autophagy-mediated ICD has not yet been elucidated. Here, we report that an oxoisoaporphine alkaloid-modified iridium(III) complex (2a) displays autophagy-inducing ICD activity. Through unbiased thermal proteome profiling (TPP), this new complex was found to interact with the lysosomal protease cathepsin D (Cat D). Subsequent cellular and biochemical assays─including the cellular thermal shift assay, isothermal dose-response assay, enzymatic assays, and molecular docking─confirmed that 2a binds to and inhibits Cat D. Further pathway analysis demonstrated that 2a triggers autophagy-dependent ICD via the LKB1-AMPK-ULK1 signaling pathway by inhibiting Cat D. Several other autophagy-dependent ICD inducers were tested and likewise found to inhibit Cat D. In contrast, an earlier reported analogue of 2a, complex 1a, was found to bind and destabilize binding immunoglobulin protein (BiP) and promote its ICD activity through an endoplasmic reticulum stress response. We believe that the findings reported here will enhance the understanding of the novel mechanisms of ICD agents and pave the way for the design of new ICD inducers with high specificity and efficacy.
Collapse
Affiliation(s)
- Yuan Lu
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
- Guangxi College Key Laboratory of Innovation Research on Medical and Engineering Integration & Liuzhou Key Laboratory of Guizhong Characteristic Medicinal Resources Development, School of Medicine, Guangxi University of Science and Technology, Liuzhou 545005, China
| | - Feng-Yang Wang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
- Key Laboratory of Medical Biotechnology and Translational Medicine, Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541004, China
| | - Matthew S Levine
- Department of Chemistry, The University of Texas at Austin, 105 E. 24th Street-A5300, Austin, Texas 78712-1224, United States
| | - Hai-Rong Shi
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Yuan Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xiaolin Xiong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou 510006, China
| | - Liang-Mei Yang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Ya-Qian Shi
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Taotao Zou
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou 510006, China
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin, 105 E. 24th Street-A5300, Austin, Texas 78712-1224, United States
| | - Hong Liang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Ke-Bin Huang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| |
Collapse
|
3
|
Nishide G, Ishibashi T, Lim K, Qiu Y, Hazawa M, Matsushima A, Wong RW. Zooming into Gene Activation: Estrogen Receptor α Dimerization and DNA Binding Visualized by High-Speed Atomic Force Microscopy. ACS NANO 2025; 19:15395-15410. [PMID: 40249907 PMCID: PMC12045019 DOI: 10.1021/acsnano.4c14943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/20/2025]
Abstract
Estrogen receptor α (ERα) is pivotal in gene regulation, particularly in estrogen-responsive cancers. However, the full-length molecular dynamic structure of ERα remains elusive. In this study, we employ high-speed atomic force microscopy (HS-AFM) to visualize ERα interactions with the estrogen response element (ERE) under both ligand-present and ligand-absent conditions. ERα binds to ERE even in the absence of estrogen, although the presence of the ligand significantly enhances binding precision and stability. Our real-time, high-resolution HS-AFM imaging captures ERα structural transitions from monomeric to dimeric forms, elucidating the molecular mechanisms by which estrogen modulates DNA-binding specificity. Based on these findings, we propose a ligand-induced dimerization (LID) model, wherein estrogen facilitates the optimal loading of ERα onto DNA. These insights deepen our understanding of hormone signaling in cancer and hold promise for the development of future therapeutic strategies targeting hormone-related malignancies.
Collapse
Affiliation(s)
- Goro Nishide
- Division
of Nano Life Science in the Graduate School of Frontier Science Initiative,
WISE Program for Nano-Precision Medicine, Science and Technology, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Tomoka Ishibashi
- Laboratory
of Structure−Function Biochemistry, Department of Chemistry,
Faculty of Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Keesiang Lim
- WPI
Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
| | - Yujia Qiu
- Division
of Nano Life Science, Graduate School of Frontier Science Initiative, Kanazawa University, Kanazawa 920-1192, Japan
| | - Masaharu Hazawa
- WPI
Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
- Cell-Bionomics
Research Unit, Innovative Integrated Bio-Research Core, Institute
for Frontier Science Initiative, Kanazawa
University, Kanazawa 920-1192, Japan
| | - Ayami Matsushima
- Laboratory
of Structure−Function Biochemistry, Department of Chemistry,
Faculty of Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Richard W. Wong
- WPI
Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
- Cell-Bionomics
Research Unit, Innovative Integrated Bio-Research Core, Institute
for Frontier Science Initiative, Kanazawa
University, Kanazawa 920-1192, Japan
| |
Collapse
|
4
|
Zhang Z, Su V, Wiese CB, Cheng L, Wang D, Cui Y, Kallapur A, Kim J, Wu X, Tran PH, Zhou Z, Casero D, Li W, Hevener AL, Reue K, Sallam T. A genome-wide ATLAS of liver chromatin accessibility reveals that sex dictates diet-induced nucleosome dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.13.623052. [PMID: 40161732 PMCID: PMC11952359 DOI: 10.1101/2024.11.13.623052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The three-dimensional organization of the genome plays an important role in cellular function. Alterations between open and closed chromatin states contributes to DNA binding, collaborative transcriptional activities and informs post-transcriptional processing. The liver orchestrates systemic metabolic control and has the ability to mount a rapid adaptive response to environmental challenges. We interrogated the chromatin architecture in liver under different dietary cues. Using ATAC-seq, we mapped over 120,000 nucleosome peaks, revealing a remarkably preserved hepatic chromatin landscape across feeding conditions. Stringent analysis of nucleosome rearrangements in response to diet revealed that sex is the dominant factor segregating changes in chromatin accessibility. A lipid-rich diet led to a more accessible chromatin confirmation at promoter regions in female mice along with enrichment of promoter binding CCAAT-binding domain proteins. Male liver exhibited stronger binding for nutrient sensing nuclear receptors. Integrative analysis with gene expression corroborated a role for chromatin states in informing functional differences in metabolic traits. We distinguished the impact of gonadal sex and chromosomal sex as determinants of chromatin modulation by diet using the Four Core Genotypes mouse model. Our data provide mechanistic evidence underlying the regulation for the critical sex-dimorphic GWAS gene, Pnpla3 . In summary, we provide a comprehensive epigenetic resource in murine liver that uncovers the complexity of chromatin dynamics in response to diet and sex. Highlights ATAC-Seq, RNA-Seq, and FCG model-integrated analysis unravel sex differences in chromatin accessibility and transcriptome responses to dietary challenges.Lipid-rich diet led to sex-biased chromatin confirmation at promoter regions.Gonadal sex emerged as the most prevalent determinant of the sex bias hepatic chromatin modulation by lipid-rich diets. The critical sex-dimorphic GWAS gene Pnpla3 is suppressed by testosterone, which underlies hepatic differences in expression between the sexes.
Collapse
|
5
|
Sivkina AL, Iarovaia OV, Razin SV, Ulianov SV. The establishment of the 3D genome structure during zygotic genome activation. Ann N Y Acad Sci 2025; 1545:38-51. [PMID: 40029160 DOI: 10.1111/nyas.15304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
During zygotic genome activation (ZGA) and early development, hierarchical levels of chromatin structure undergo remarkable perturbations: changes in the nuclear-to-cytoplasmic ratio of various components; changes in chromatin accessibility; histone exchange; and the formation of 3D structures such as loops, topologically associated domains, and compartments. Here, we review the peculiarities, variability, and emergence of the chromatin structural features during ZGA in different organisms. Focusing on newly found structures called fountains, we describe the prerequisites for cohesin loading on DNA and possible mechanisms of genome organization in early development. Fountains resulting from asymmetric bidirectional cohesin extrusion spread from cohesin-loading points in a CTCF-independent manner. We discuss that fountains may not possess specific functions, unlike conventional chromatin structures, and could be found in other biological processes where cohesin loading occurs.
Collapse
Affiliation(s)
| | - Olga V Iarovaia
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Sergey V Razin
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Sergey V Ulianov
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
6
|
Meli R, Aksoy O, Vallet S, Slade D, Podar K. Can we develop effective direct or indirect inhibitors of transcription factors? On the clinical evolution of protein degraders for multiple myeloma therapy. Expert Opin Ther Targets 2025; 29:101-115. [PMID: 40122131 DOI: 10.1080/14728222.2025.2482557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Transcription factors (TFs) are master regulators of cellular function and orchestrate diverse signaling pathways and processes. Acting as convergence points of signaling pathways, they integrate extracellular stimuli with intracellular responses to regulate cell functions. Dysregulation of TFs drives tumorigenesis including proliferation, drug resistance, and immune evasion of multiple myeloma (MM), the second most-common hematologic malignancy. AREAS COVERED The discovery that IMiDs are molecular glue degraders, which reprogram the E3-ligase cereblon (CRBN) to ubiquitinate and degrade IKZF1 and IKZF3, two otherwise un-druggable crucial TFs in MM, gave rise to the widespread interest in proximity-induced protein-degradation as an exciting novel therapeutic strategy. This review summarizes our up-to-date knowledge on the pre/clinical development of IMiD-related, more potent CRBN E3-Ligase Modulatory Drugs (CELMoDs), directed PROteolysis TArgeting Chimeras (PROTACs) and degronomids as well as on promising future avenues in the field of targeted protein-degradation (TPD). EXPERT OPINION TPD is an emerging field to treat cancer, including MM. CELMoDs are already reshaping the treatment landscape of MM. Preclinical data on PROTACs are promising. Nevertheless, a deeper understanding of TF biology as well as further advancements in screening methodologies and chemoproteomics are crucial to further spur the transformative potential of targeted TF degradation in MM.
Collapse
Affiliation(s)
- Rajeshwari Meli
- Division of Molecular Oncology and Hematology, Department of Basic and Translational Oncology, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| | - Osman Aksoy
- Division of Molecular Oncology and Hematology, Department of Basic and Translational Oncology, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| | - Sonia Vallet
- Division of Molecular Oncology and Hematology, Department of Basic and Translational Oncology, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
- Division of Internal Medicine, University Hospital Krems, Krems an der Donau, Austria
| | - Dea Slade
- MedAustron Ion Therapy Center, Wiener Neustadt, Austria
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Center for Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Klaus Podar
- Division of Molecular Oncology and Hematology, Department of Basic and Translational Oncology, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
- Division of Internal Medicine, University Hospital Krems, Krems an der Donau, Austria
| |
Collapse
|
7
|
Zhao Q. Thermodynamic for biological development: A hypothesis. Biosystems 2025; 249:105413. [PMID: 39929432 DOI: 10.1016/j.biosystems.2025.105413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
This paper proposes a thermodynamic model of biological development. Several key thoughts are presented: 1) in view of thermodynamics, biological development processes irreversibly; 2) in view of thermodynamics and molecular biology, positive autoregulation, or self-regulation, of transcription factors is the only way to ensure irreversibility of a thermodynamic process of biology; 3) change in the autoregulation of transcription factors can irreversibly result in alterations in the physiological state) a physiological state is a system of signaling networks; 5) a cell and its physiological state can be identified by the pattern of its transcription factors. 6) from points aforementioned, we can analyze some thermodynamic properties of biological development by knowledge of molecular biology and biochemistry. The possible mechanisms of plant vernalization are also proposed.
Collapse
Affiliation(s)
- Qinyi Zhao
- Medical Institute, CRRC, Beijing, PR China.
| |
Collapse
|
8
|
Wan B, Yu J. Protein target search diffusion-association/dissociation free energy landscape around DNA binding site with flanking sequences. Biophys J 2025; 124:677-692. [PMID: 39818622 PMCID: PMC11900189 DOI: 10.1016/j.bpj.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/05/2024] [Accepted: 01/13/2025] [Indexed: 01/18/2025] Open
Abstract
In this work we present a minimal structure-based model of protein diffusional search along local DNA amid protein binding and unbinding events on the DNA, taking into account protein-DNA electrostatic interactions and hydrogen-bonding (HB) interactions or contacts at the interface. We accordingly constructed the protein diffusion-association/dissociation free energy surface and mapped it to 1D as the protein slides along DNA, maintaining the protein-DNA interfacial HB contacts that presumably dictate the DNA sequence information detection. Upon DNA helical path correction, the protein 1D diffusion rates along local DNA can be physically derived to be consistent with experimental measurements. We also show that the sequence-dependent protein sliding or stepping patterns along DNA are regulated by collective interfacial HB dynamics, which also determines the ruggedness of the protein diffusion free energy landscape on the local DNA. In comparison, protein association or binding with DNA are generically dictated by the protein-DNA electrostatic interactions, with an interaction zone of nanometers around DNA. Extra degrees of freedom (DOFs) of the protein such as rotations and conformational fluctuations can be well accommodated within the protein-DNA electrostatic interaction zone. As such we demonstrate that the protein binding or association free energy profiling along DNA smoothens over the 1D diffusion free energy landscape, which leads to population variations for an order of magnitude upon a marginal free energetic smoothening around the specific or consensus sites. We further show that the protein unbinding or dissociation from a comparatively high-binding affinity DNA site is dominated by lateral diffusion to the flanking low-affinity sites. The results predict that experimental characterizations on the relative protein-DNA binding affinities or population profiling on the DNA are systematically and physically impacted by the extra DOFs of protein motions aside from 1D translation or helical tracking, as well as from flanking DNA sequences due to protein 1D diffusion and nonspecific binding/unbinding.
Collapse
Affiliation(s)
- Biao Wan
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, China
| | - Jin Yu
- Department of Physics and Astronomy, Department of Chemistry, NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California.
| |
Collapse
|
9
|
Raveh B, Eliasian R, Rashkovits S, Russel D, Hayama R, Sparks S, Singh D, Lim R, Villa E, Rout MP, Cowburn D, Sali A. Integrative mapping reveals molecular features underlying the mechanism of nucleocytoplasmic transport. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.31.573409. [PMID: 38260487 PMCID: PMC10802240 DOI: 10.1101/2023.12.31.573409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Nuclear Pore Complexes (NPCs) enable rapid, selective, and robust nucleocytoplasmic transport. To explain how transport emerges from the system components and their interactions, we used experimental data and theoretical information to construct an integrative Brownian dynamics model of transport through an NPC, coupled to a kinetic model of transport in the cell. The model recapitulates key aspects of transport for a wide range of molecular cargos, including pre-ribosomes and viral capsids. It quantifies how flexible phenylalanine-glycine (FG) repeat proteins raise an entropy barrier to passive diffusion and how this barrier is selectively lowered in facilitated diffusion by the many transient interactions of nuclear transport receptors with the FG repeats. Selective transport is enhanced by "fuzzy" multivalent interactions, redundant FG repeats, coupling to the energy-dependent RanGTP concentration gradient, and exponential dependence of transport kinetics on the transport barrier. Our model will facilitate rational modulation of the NPC and its artificial mimics.
Collapse
|
10
|
O'Dwyer MR, Azagury M, Furlong K, Alsheikh A, Hall-Ponsele E, Pinto H, Fyodorov DV, Jaber M, Papachristoforou E, Benchetrit H, Ashmore J, Makedonski K, Rahamim M, Hanzevacki M, Yassen H, Skoda S, Levy A, Pollard SM, Skoultchi AI, Buganim Y, Soufi A. Nucleosome fibre topology guides transcription factor binding to enhancers. Nature 2025; 638:251-260. [PMID: 39695228 PMCID: PMC11798873 DOI: 10.1038/s41586-024-08333-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
Cellular identity requires the concerted action of multiple transcription factors (TFs) bound together to enhancers of cell-type-specific genes. Despite TFs recognizing specific DNA motifs within accessible chromatin, this information is insufficient to explain how TFs select enhancers1. Here we compared four different TF combinations that induce different cell states, analysing TF genome occupancy, chromatin accessibility, nucleosome positioning and 3D genome organization at the nucleosome resolution. We show that motif recognition on mononucleosomes can decipher only the individual binding of TFs. When bound together, TFs act cooperatively or competitively to target nucleosome arrays with defined 3D organization, displaying motifs in particular patterns. In one combination, motif directionality funnels TF combinatorial binding along chromatin loops, before infiltrating laterally to adjacent enhancers. In other combinations, TFs assemble on motif-dense and highly interconnected loop junctions, and subsequently translocate to nearby lineage-specific sites. We propose a guided-search model in which motif grammar on nucleosome fibres acts as signpost elements, directing TF combinatorial binding to enhancers.
Collapse
Affiliation(s)
- Michael R O'Dwyer
- Institute of Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Institute of Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Meir Azagury
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Katharine Furlong
- Institute of Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Institute of Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh, UK
| | - Amani Alsheikh
- Institute of Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Institute of Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
- Health Sector, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Elisa Hall-Ponsele
- Institute of Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Institute of Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Hugo Pinto
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, USA
| | - Dmitry V Fyodorov
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, USA
| | - Mohammad Jaber
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Eleni Papachristoforou
- Institute of Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Hana Benchetrit
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - James Ashmore
- Institute of Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Kirill Makedonski
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Moran Rahamim
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Marta Hanzevacki
- Institute of Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Institute of Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Hazar Yassen
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Samuel Skoda
- Institute of Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Institute of Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Adi Levy
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Steven M Pollard
- Institute of Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh, UK
| | - Arthur I Skoultchi
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, USA
| | - Yosef Buganim
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| | - Abdenour Soufi
- Institute of Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK.
- Institute of Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK.
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
11
|
Huynh D, Hoffmeister P, Friedrich T, Zhang K, Bartkuhn M, Ferrante F, Giaimo BD, Kovall RA, Borggrefe T, Oswald F, Gebhardt JCM. Effective in vivo binding energy landscape illustrates kinetic stability of RBPJ-DNA binding. Nat Commun 2025; 16:1259. [PMID: 39893191 PMCID: PMC11787368 DOI: 10.1038/s41467-025-56515-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 01/21/2025] [Indexed: 02/04/2025] Open
Abstract
Transcription factors (TFs) such as RBPJ in Notch signaling bind to specific DNA sequences to regulate transcription. How TF-DNA binding kinetics and cofactor interactions modulate gene regulation is mostly unknown. We determine the binding kinetics, transcriptional activity, and genome-wide chromatin occupation of RBPJ and mutant variants by live-cell single-molecule tracking, reporter assays, and ChIP-Seq. Importantly, the search time of RBPJ exceeds its residence time, indicating kinetic rather than thermodynamic binding stability. Impaired RBPJ-DNA binding as in Adams-Oliver-Syndrome affect both target site association and dissociation, while impaired cofactor binding mainly alters association and unspecific binding. Moreover, our data point to the possibility that cofactor binding contributes to target site specificity. Findings for other TFs comparable to RBPJ indicate that kinetic rather than thermodynamic DNA binding stability might prevail in vivo. We propose an effective in vivo binding energy landscape of TF-DNA interactions as instructive visualization of binding kinetics and mutation-induced changes.
Collapse
Affiliation(s)
- Duyen Huynh
- Institute of Experimental Physics and IQST, Ulm University, Ulm, Germany
| | | | - Tobias Friedrich
- Institute of Biochemistry, Justus-Liebig-Universität Gießen, Gießen, Germany
- Biomedical Informatics and Systems Medicine, Justus-Liebig-Universität Gießen, Gießen, Germany
- Institute for Lung Health (ILH), Gießen, Germany
| | - Kefan Zhang
- Institute of Experimental Physics and IQST, Ulm University, Ulm, Germany
| | - Marek Bartkuhn
- Biomedical Informatics and Systems Medicine, Justus-Liebig-Universität Gießen, Gießen, Germany
- Institute for Lung Health (ILH), Gießen, Germany
| | - Francesca Ferrante
- Institute of Biochemistry, Justus-Liebig-Universität Gießen, Gießen, Germany
| | | | - Rhett A Kovall
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tilman Borggrefe
- Institute of Biochemistry, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Franz Oswald
- Clinic of Internal Medicine I, University Medical Center Ulm, Ulm, Germany.
| | | |
Collapse
|
12
|
Wang K, Liu S, Zhou S, Qileng A, Wang D, Liu Y, Chen C, Lei C, Nie Z. Ligand-Responsive Artificial Protein-Protein Communication for Field-Deployable Cell-Free Biosensing. Angew Chem Int Ed Engl 2025; 64:e202416671. [PMID: 39558180 DOI: 10.1002/anie.202416671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/20/2024]
Abstract
Natural protein-protein communications, such as those between transcription factors (TFs) and RNA polymerases/ribosomes, underpin cell-free biosensing systems operating on the transcription/translation (TXTL) paradigm. However, their deployment in field analysis is hampered by the delayed response (hour-level) and the complex composition of in vitro TXTL systems. For this purpose, we present a de novo-designed ligand-responsive artificial protein-protein communication (LIRAC) by redefining the connection between TFs and non-interacting CRISPR/Cas enzymes. By rationally designing a chimeric DNA adaptor and precisely regulating its binding affinities to both proteins, LIRAC immediately transduces target-induced TF allostery into rapid CRISPR/Cas enzyme activation within a homogeneous system. Consequently, LIRAC obviates the need for RNA/protein biosynthesis inherent to conventional TXTL-based cell-free systems, substantially reducing reaction complexity and time (from hours to 10 minutes) with improved sensitivity and tunable dynamic range. Moreover, LIRAC exhibits excellent versatility and programmability for rapidly and sensitively detecting diverse contaminants, including antibiotics, heavy metal ions, and preservatives. It also enables the creation of a multi-protein communication-based tristate logic for the intelligent detection of multiple contaminants. Integrated with portable devices, LIRAC has been proven effective in the field analysis of environmental samples and personal care products, showcasing its potential for environmental and health monitoring.
Collapse
Affiliation(s)
- Ke Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, 410082, P. R. China
| | - Siqian Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, 410082, P. R. China
| | - Shuqi Zhou
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, 100084, P. R. China
| | - Aori Qileng
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, P. R. China
| | - Dingyi Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
| | - Yingju Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, P. R. China
| | - Chunlai Chen
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, 100084, P. R. China
| | - Chunyang Lei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, 410082, P. R. China
| | - Zhou Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, 410082, P. R. China
| |
Collapse
|
13
|
Wagh K, Stavreva DA, Hager GL. Transcription dynamics and genome organization in the mammalian nucleus: Recent advances. Mol Cell 2025; 85:208-224. [PMID: 39413793 PMCID: PMC11741928 DOI: 10.1016/j.molcel.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/31/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024]
Abstract
Single-molecule tracking (SMT) has emerged as the dominant technology to investigate the dynamics of chromatin-transcription factor (TF) interactions. How long a TF needs to bind to a regulatory site to elicit a transcriptional response is a fundamentally important question. However, highly divergent estimates of TF binding have been presented in the literature, stemming from differences in photobleaching correction and data analysis. TF movement is often interpreted as specific or non-specific association with chromatin, yet the dynamic nature of the chromatin polymer is often overlooked. In this perspective, we highlight how recent SMT studies have reshaped our understanding of TF dynamics, chromatin mobility, and genome organization in the mammalian nucleus, focusing on the technical details and biological implications of these approaches. In a remarkable convergence of fixed and live-cell imaging, we show how super-resolution and SMT studies of chromatin have dovetailed to provide a convincing nanoscale view of genome organization.
Collapse
Affiliation(s)
- Kaustubh Wagh
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diana A Stavreva
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
14
|
Hastings R, Aditham AK, DelRosso N, Suzuki PH, Fordyce PM. Mutations to transcription factor MAX allosterically increase DNA selectivity by altering folding and binding pathways. Nat Commun 2025; 16:636. [PMID: 39805837 PMCID: PMC11729911 DOI: 10.1038/s41467-024-55672-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
Understanding how proteins discriminate between preferred and non-preferred ligands ('selectivity') is essential for predicting biological function and a central goal of protein engineering efforts, yet the biophysical mechanisms underpinning selectivity remain poorly understood. Towards this end, we study how variants of the promiscuous transcription factor (TF) MAX (H. sapiens) alter DNA specificity and selectivity, yielding >1700 Kds and >500 rate constants in complex with multiple DNA sequences. Twenty-two of the 240 assayed MAX point mutations enhance selectivity, yet none of these mutations occur at residues that contact nucleotides in published structures. By applying thermodynamic and kinetic models to these results and previous observations for the highly similar yet far more selective TF Pho4 (S. cerevisiae), we find that these mutations enhance selectivity by altering partitioning between or affinity within conformations with different intrinsic selectivity, providing a mechanistic basis for allosteric modulation of ligand selectivity. These results highlight the importance of conformational heterogeneity in determining sequence selectivity and can guide future efforts to engineer selective proteins.
Collapse
Affiliation(s)
- Renee Hastings
- Biophysics Program, Stanford University, Stanford, CA, USA
| | - Arjun K Aditham
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | | | - Peter H Suzuki
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Polly M Fordyce
- Biophysics Program, Stanford University, Stanford, CA, USA.
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
15
|
Jyoti, Ritu, Gupta S, Shankar R. Comprehensive analysis of computational approaches in plant transcription factors binding regions discovery. Heliyon 2024; 10:e39140. [PMID: 39640721 PMCID: PMC11620080 DOI: 10.1016/j.heliyon.2024.e39140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/23/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
Transcription factors (TFs) are regulatory proteins which bind to a specific DNA region known as the transcription factor binding regions (TFBRs) to regulate the rate of transcription process. The identification of TFBRs has been made possible by a number of experimental and computational techniques established during the past few years. The process of TFBR identification involves peak identification in the binding data, followed by the identification of motif characteristics. Using the same binding data attempts have been made to raise computational models to identify such binding regions which could save time and resources spent for binding experiments. These computational approaches depend a lot on what way they learn and how. These existing computational approaches are skewed heavily around human TFBRs discovery, while plants have drastically different genomic setup for regulation which these approaches have grossly ignored. Here, we provide a comprehensive study of the current state of the matters in plant specific TF discovery algorithms. While doing so, we encountered several software tools' issues rendering the tools not useable to researches. We fixed them and have also provided the corrected scripts for such tools. We expect this study to serve as a guide for better understanding of software tools' approaches for plant specific TFBRs discovery and the care to be taken while applying them, especially during cross-species applications. The corrected scripts of these software tools are made available at https://github.com/SCBB-LAB/Comparative-analysis-of-plant-TFBS-software.
Collapse
Affiliation(s)
- Jyoti
- Studio of Computational Biology & Bioinformatics, The Himalayan Centre for High-throughput Computational Biology, (HiCHiCoB, A BIC Supported by DBT, India), Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, (HP), 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Ritu
- Studio of Computational Biology & Bioinformatics, The Himalayan Centre for High-throughput Computational Biology, (HiCHiCoB, A BIC Supported by DBT, India), Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, (HP), 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Sagar Gupta
- Studio of Computational Biology & Bioinformatics, The Himalayan Centre for High-throughput Computational Biology, (HiCHiCoB, A BIC Supported by DBT, India), Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, (HP), 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Ravi Shankar
- Studio of Computational Biology & Bioinformatics, The Himalayan Centre for High-throughput Computational Biology, (HiCHiCoB, A BIC Supported by DBT, India), Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, (HP), 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
16
|
Shen L, Bai X, Zhao L, Zhou J, Chang C, Li X, Cao Z, Li Y, Luan P, Li H, Zhang H. Integrative 3D genomics with multi-omics analysis and functional validation of genetic regulatory mechanisms of abdominal fat deposition in chickens. Nat Commun 2024; 15:9274. [PMID: 39468045 PMCID: PMC11519623 DOI: 10.1038/s41467-024-53692-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024] Open
Abstract
Chickens are the most abundant agricultural animals globally, with controlling abdominal fat deposition being a key objective in poultry breeding. While GWAS can identify genetic variants associated with abdominal fat deposition, the precise roles and mechanisms of these variants remain largely unclear. Here, we use male chickens from two lines divergently selected for abdominal fat deposition as experimental models. Through the integration of genomic, epigenomic, 3D genomic, and transcriptomic data, we build a comprehensive chromatin 3D regulatory network map to identify the genetic regulatory mechanisms that influence abdominal fat deposition in chickens. Notably, we find that the rs734209466 variant functions as an allele-specific enhancer, remotely enhancing the transcription of IGFBP2 and IGFBP5 by the binding transcription factor IRF4. This interaction influences the differentiation and proliferation of preadipocytes, which ultimately affects phenotype. This work presents a detailed genetic regulatory map for chicken abdominal fat deposition, offering molecular targets for selective breeding.
Collapse
Affiliation(s)
- Linyong Shen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, PR China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, PR China
| | - Xue Bai
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, PR China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, PR China
| | - Liru Zhao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, PR China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, PR China
| | - Jiamei Zhou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, PR China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, PR China
| | - Cheng Chang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, PR China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, PR China
| | - Xinquan Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, PR China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, PR China
| | - Zhiping Cao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, PR China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, PR China
| | - Yumao Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, PR China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, PR China
| | - Peng Luan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, PR China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, PR China
| | - Hui Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China.
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, PR China.
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, PR China.
| | - Hui Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China.
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, PR China.
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, PR China.
| |
Collapse
|
17
|
Rauluseviciute I, Launay T, Barzaghi G, Nikumbh S, Lenhard B, Krebs AR, Castro-Mondragon J, Mathelier A. Identification of transcription factor co-binding patterns with non-negative matrix factorization. Nucleic Acids Res 2024; 52:e85. [PMID: 39217462 PMCID: PMC11472169 DOI: 10.1093/nar/gkae743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/12/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Transcription factor (TF) binding to DNA is critical to transcription regulation. Although the binding properties of numerous individual TFs are well-documented, a more detailed comprehension of how TFs interact cooperatively with DNA is required. We present COBIND, a novel method based on non-negative matrix factorization (NMF) to identify TF co-binding patterns automatically. COBIND applies NMF to one-hot encoded regions flanking known TF binding sites (TFBSs) to pinpoint enriched DNA patterns at fixed distances. We applied COBIND to 5699 TFBS datasets from UniBind for 401 TFs in seven species. The method uncovered already established co-binding patterns and new co-binding configurations not yet reported in the literature and inferred through motif similarity and protein-protein interaction knowledge. Our extensive analyses across species revealed that 67% of the TFs shared a co-binding motif with other TFs from the same structural family. The co-binding patterns captured by COBIND are likely functionally relevant as they harbor higher evolutionarily conservation than isolated TFBSs. Open chromatin data from matching human cell lines further supported the co-binding predictions. Finally, we used single-molecule footprinting data from mouse embryonic stem cells to confirm that the COBIND-predicted co-binding events associated with some TFs likely occurred on the same DNA molecules.
Collapse
Affiliation(s)
- Ieva Rauluseviciute
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
| | - Timothée Launay
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
| | - Guido Barzaghi
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany
- Collaboration for Joint Ph.D. degree between EMBL and Heidelberg University, Heidelberg, Germany
| | - Sarvesh Nikumbh
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Boris Lenhard
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Arnaud Regis Krebs
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Jaime A Castro-Mondragon
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
| | - Anthony Mathelier
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
- Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Center for Bioinformatics, Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
18
|
Flora P, Li MY, Zhou Y, Mercédes M, Zheng XY, Galbo PM, Zheng D, Ezhkova E. H2AK119ub dynamics controls hair follicle stem cell quiescence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617646. [PMID: 39416158 PMCID: PMC11482967 DOI: 10.1101/2024.10.10.617646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The transition of stem cells from a quiescent state to an active state is a finely tuned process that requires the dismantling of the quiescence program and the establishment of a cell cycle-promoting transcriptional landscape. Whether epigenetic processes control stem cell states to promote the regeneration of adult tissues remains elusive. In this study, we show that a repressive histone modification, H2AK119ub, is dynamic between quiescent and active hair follicle stem cells (HFSCs) in the adult murine skin. Ablation of H2AK119ub in HFSCs leads to impaired quiescence leading to premature activation and an eventual exhaustion of HFSC pool. Transcriptional and chromatin studies revealed that H2AK119ub directly represses a proliferation promoting transcriptional program in the HFSCs to preserve quiescence. Lastly, we identify that the inhibitory FGF signaling produced by the hair follicle niche keratinocytes maintains H2AK119ub in quiescent HFSCs. Together, these findings reveal that a repressive histone mark, H2AK119ub, is under the dynamic regulation of inhibitory niche signaling to prevent the untimely establishment of an activated state to preserve SC function and longevity.
Collapse
|
19
|
Perumal CM, Thulo M, Buthelezi S, Naicker P, Stoychev S, Lakhi A, Fanucchi S. Unraveling the interplay between the leucine zipper and forkhead domains of FOXP2: Implications for DNA binding, stability and dynamics. Proteins 2024; 92:1177-1189. [PMID: 38747678 DOI: 10.1002/prot.26699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/21/2024] [Accepted: 04/26/2024] [Indexed: 10/26/2024]
Abstract
FOXP2 is a transcription factor associated with speech and language. Like other FOX transcription factors, it has a DNA binding region called the forkhead domain (FHD). This domain can exist as a monomer or a domain swapped dimer. In addition to the FHD, the leucine zipper region (LZ) of FOXP2 is also believed to be associated with both DNA binding and oligomerization. To better understand the relationship between DNA binding and oligomerization of FOXP2, we investigated its structure, stability and dynamics, focusing specifically on the FHD and the LZ. We did this by using two constructs: one containing the isolated FHD and one containing both the LZ and the FHD (LZ-END). We demonstrate in this work, that while the FHD maintains a monomeric form that is capable of binding DNA, the LZ-END undergoes a dynamic transition between oligomeric states in the presence of DNA. Our findings suggest that FOXP2's LZ domain influences DNA binding affinity through a change in oligomeric state. We show through hydrogen exchange mass spectroscopy that certain parts of the FHD and interlinking region become less dynamic when in the presence of DNA, confirming DNA binding and oligomerization in these regions. Moreover, the detection of a stable equilibrium intermediate state during LZ-END unfolding supports the idea of cooperation between these two domains. Overall, our study sheds light on the interplay between two FOXP2 domains, providing insight into the protein's ability to respond dynamically to DNA, and enriching our understanding of FOXP2's role in gene regulation.
Collapse
Affiliation(s)
- Cardon Maria Perumal
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Monare Thulo
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | | | | | | | - Aasiya Lakhi
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Sylvia Fanucchi
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| |
Collapse
|
20
|
Carminati M, Vecchia L, Stoos L, Thomä NH. Pioneer factors: Emerging rules of engagement for transcription factors on chromatinized DNA. Curr Opin Struct Biol 2024; 88:102875. [PMID: 38991237 DOI: 10.1016/j.sbi.2024.102875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 07/13/2024]
Abstract
Pioneering transcription factors (TFs) can drive cell fate changes by binding their DNA motifs in a repressive chromatin environment. Recent structures illustrate emerging rules for nucleosome engagement: TFs distort the nucleosomal DNA to gain access or employ alternative DNA-binding modes with smaller footprints, they preferentially access solvent-exposed motifs near the entry/exit sites, and frequently interact with histones. The extent of TF-histone interactions, in turn, depends on the motif location on the nucleosome, the type of DNA-binding fold, and adjacent domains present. TF-histone interactions can phase TF motifs relative to nucleosomes, and we discuss how these complex and surprisingly diverse interactions between nucleosomes and TFs contribute to function.
Collapse
Affiliation(s)
- Manuel Carminati
- Swiss Institute for Experimental Cancer Research (ISREC), EPFL, Lausanne 1015, Switzerland
| | - Luca Vecchia
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel 4058, Switzerland
| | - Lisa Stoos
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel 4058, Switzerland
| | - Nicolas H Thomä
- Swiss Institute for Experimental Cancer Research (ISREC), EPFL, Lausanne 1015, Switzerland; Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel 4058, Switzerland.
| |
Collapse
|
21
|
Chen JH, Li JJ, Yuan Y, Tian Q, Feng DD, Zhuang LL, Cao Q, Zhou GP, Jin R. ETS1 and RBPJ transcriptionally regulate METTL14 to suppress TGF-β1-induced epithelial-mesenchymal transition in human bronchial epithelial cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167349. [PMID: 39002703 DOI: 10.1016/j.bbadis.2024.167349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
Asthma is a chronic respiratory disease characterized by airway inflammation and remodeling. Epithelial-mesenchymal transition (EMT) of bronchial epithelial cells is considered to be a crucial player in asthma. Methyltransferase-like 14 (METTL14), an RNA methyltransferase, is implicated in multiple pathological processes, including EMT, cell proliferation and migration. However, the role of METTL14 in asthma remains uncertain. This research aimed to explore the biological functions of METTL14 in asthma and its underlying upstream mechanisms. METTL14 expression was down-regulated in asthmatic from three GEO datasets (GSE104468, GSE165934, and GSE74986). Consistent with this trend, METTL14 was decreased in the lung tissues of OVA-induced asthmatic mice and transforming growth factor-β1 (TGF-β1)-stimulated human bronchial epithelial cells (Beas-2B) in this study. Overexpression of METTL14 caused reduction in mesenchymal markers (FN1, N-cad, Col-1 and α-SMA) in TGF-β1-treated cells, but caused increase in epithelial markers (E-cad), thus inhibiting EMT. Also, METTL14 suppressed the proliferation and migration ability of TGF-β1-treated Beas-2B cells. Two transcription factors, ETS1 and RBPJ, could both bind to the promoter region of METTL14 and drive its expression. Elevating METTL14 expression could reversed EMT, cell proliferation and migration promoted by ETS1 or RBPJ deficiency. These results indicate that the ETS1/METTL14 and RBPJ/METTL14 transcription axes exhibit anti-EMT, anti-proliferation and anti-migration functions in TGF-β1-induced bronchial epithelial cells, implying that METTL14 may be considered an alternative candidate target for the treatment of asthma.
Collapse
Affiliation(s)
- Jia-He Chen
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Jiao-Jiao Li
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Yue Yuan
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Qiang Tian
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Dan-Dan Feng
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Li-Li Zhuang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Qian Cao
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| | - Guo-Ping Zhou
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China; Clinical Allergy Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| | - Rui Jin
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China; Clinical Allergy Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
22
|
Chen D, Zhao J, Xu S, Wu L. Detection of Short-Chain Chlorinated Aliphatic Hydrocarbons through an Engineered Biosensor with Tailored Ligand Specificity. Anal Chem 2024; 96:15614-15623. [PMID: 39292503 DOI: 10.1021/acs.analchem.4c02476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Short-chain chlorinated aliphatic hydrocarbons (SCAHs), commonly used as industrial reagents and solvents, pose a significant threat to ecosystems and human health as they infiltrate aquatic environments due to extensive usage and accidental spills. Whole-cell biosensors have emerged as cost-effective, rapid, and real-time analytical tools for environmental monitoring and remediation. While the broad ligand specificity of transcriptional factors (TFs) often prohibits the application of such biosensors. Herein, we exploited a semirational transition ligand approach in conjunction with a positive/negative fluorescence-activated cell sorting (FACS) strategy to develop a biosensor based on the TF AlkS, which is highly specific for SCAHs. Furthermore, through promoter-directed evolution, the performance of the biosensor was further enhanced. Mutation in the -10 region of constitutive promoter PalkS resulted in reduced AlkS leakage expression, while mutation in the -10 region of inducible promoter PalkB increased its accessibility to the AlkS-SCAHs complex. This led to an 89% reduction in background fluorescence leakage of the optimized biosensor, M2-463, further enhancing its response to SCAHs. The optimized biosensor was highly sensitive and exhibited a broader dynamic response range with a 150-fold increase in fluorescence output after 1 h of induction. The detection limit (LOD) reached 0.03 ppm, and the average recovery rate of SCAHs in actual water samples ranged from 95.87 to 101.20%. The accuracy and precision of the proposed biosensor were validated using gas chromatography-mass spectrometry (GC-MS), demonstrating the promising application for SCAH detection in an actual environment sample.
Collapse
Affiliation(s)
- Dongdong Chen
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Jiadi Zhao
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Shengmin Xu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Lijun Wu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| |
Collapse
|
23
|
Luo Z, Wu L, Miao X, Zhang S, Wei N, Zhao S, Shang X, Hu H, Xue J, Zhang T, Yang F, Xu S, Li L. A dynamic regulome of shoot-apical-meristem-related homeobox transcription factors modulates plant architecture in maize. Genome Biol 2024; 25:245. [PMID: 39300560 DOI: 10.1186/s13059-024-03391-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND The shoot apical meristem (SAM), from which all above-ground tissues of plants are derived, is critical to plant morphology and development. In maize (Zea mays), loss-of-function mutant studies have identified several SAM-related genes, most encoding homeobox transcription factors (TFs), located upstream of hierarchical networks of hundreds of genes. RESULTS Here, we collect 46 transcriptome and 16 translatome datasets across 62 different tissues or stages from the maize inbred line B73. We construct a dynamic regulome for 27 members of three SAM-related homeobox subfamilies (KNOX, WOX, and ZF-HD) through machine-learning models for the detection of TF targets across different tissues and stages by combining tsCUT&Tag, ATAC-seq, and expression profiling. This dynamic regulome demonstrates the distinct binding specificity and co-factors for these homeobox subfamilies, indicative of functional divergence between and within them. Furthermore, we assemble a SAM dynamic regulome, illustrating potential functional mechanisms associated with plant architecture. Lastly, we generate a wox13a mutant that provides evidence that WOX13A directly regulates Gn1 expression to modulate plant height, validating the regulome of SAM-related homeobox genes. CONCLUSIONS The SAM-related homeobox transcription-factor regulome presents an unprecedented opportunity to dissect the molecular mechanisms governing SAM maintenance and development, thereby advancing our understanding of maize growth and shoot architecture.
Collapse
Affiliation(s)
- Zi Luo
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Leiming Wu
- The National Engineering Laboratory of Crop Resistance Breeding, School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Xinxin Miao
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shuang Zhang
- The Key Laboratory of Biology and Genetics Improvement of Maize in Arid Area of Northwest Region, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, 712199, China
| | - Ningning Wei
- The Key Laboratory of Biology and Genetics Improvement of Maize in Arid Area of Northwest Region, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, 712199, China
| | - Shiya Zhao
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiaoyang Shang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hongyan Hu
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jiquan Xue
- The Key Laboratory of Biology and Genetics Improvement of Maize in Arid Area of Northwest Region, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, 712199, China
| | - Tifu Zhang
- Jiangsu Provincial Key Laboratory of Agrobiology, Institute of Germplasm Resources and Biotechnology, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Fang Yang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shutu Xu
- The Key Laboratory of Biology and Genetics Improvement of Maize in Arid Area of Northwest Region, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, 712199, China.
| | - Lin Li
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
- Hubei Hongshan Laboratory, Wuhan, 430070, China.
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan, 430070, China.
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China.
| |
Collapse
|
24
|
Dhillon N, Kamakaka RT. Transcriptional silencing in Saccharomyces cerevisiae: known unknowns. Epigenetics Chromatin 2024; 17:28. [PMID: 39272151 PMCID: PMC11401328 DOI: 10.1186/s13072-024-00553-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Transcriptional silencing in Saccharomyces cerevisiae is a persistent and highly stable form of gene repression. It involves DNA silencers and repressor proteins that bind nucleosomes. The silenced state is influenced by numerous factors including the concentration of repressors, nature of activators, architecture of regulatory elements, modifying enzymes and the dynamics of chromatin.Silencers function to increase the residence time of repressor Sir proteins at silenced domains while clustering of silenced domains enables increased concentrations of repressors and helps facilitate long-range interactions. The presence of an accessible NDR at the regulatory regions of silenced genes, the cycling of chromatin configurations at regulatory sites, the mobility of Sir proteins, and the non-uniform distribution of the Sir proteins across the silenced domain, all result in silenced chromatin that only stably silences weak promoters and enhancers via changes in transcription burst duration and frequency.These data collectively suggest that silencing is probabilistic and the robustness of silencing is achieved through sub-optimization of many different nodes of action such that a stable expression state is generated and maintained even though individual constituents are in constant flux.
Collapse
Affiliation(s)
- Namrita Dhillon
- Department of Biomolecular Engineering, University of California, 1156 High Street, Santa Cruz, CA, 95064, USA
| | - Rohinton T Kamakaka
- Department of MCD Biology, University of California, 1156 High Street, Santa Cruz, CA, 95064, USA.
| |
Collapse
|
25
|
Wang G, Wu Y, Su Y, Qu N, Chen B, Zhou D, Yuan L, Yin M, Liu M, Zhou W. TCF12-regulated GRB7 facilitates the HER2+ breast cancer progression by activating Notch1 signaling pathway. J Transl Med 2024; 22:745. [PMID: 39113057 PMCID: PMC11304905 DOI: 10.1186/s12967-024-05536-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 07/24/2024] [Indexed: 08/11/2024] Open
Abstract
BACKGROUND Human epidermal growth factor receptor 2-positive (HER2+) breast cancer (BC), which accounts for approximately one-fifth of all BCs, are highly invasive with a high rate of recurrence and a poor prognosis. Several studies have shown that growth factor receptor-bound protein 7 (GRB7) might be a potential therapeutic target for tumor diagnosis and prognosis. Nevertheless, the role of GRB7 in HER2+ BC and its underlying mechanisms have not been fully elucidated. The aim of this study was to investigate the biological function and regulatory mechanism of GRB7 in HER2+ BC. METHODS Bioinformatics analysis was performed using the TCGA, GEO and CancerSEA databases to evaluate the clinical significance of GRB7. RT quantitative PCR, western blot and immunofluorescence were conducted to assess the expression of GRB7 in BC cell lines and tissues. MTT, EdU, colony formation, wound healing, transwell, and xenograft assays were adopted to explore the biological function of GRB7 in HER2+ BC. RNA sequencing was performed to analyze the signaling pathways associated with GRB7 in SK-BR-3 cells after the cells were transfected with GRB7 siRNA. Chromatin immunoprecipitation analysis (ChIP) and luciferase reporter assay were employed to elucidate the potential molecular regulatory mechanisms of GRB7 in HER2+ BC. RESULTS GRB7 was markedly upregulated and associated with poor prognosis in BC, especially in HER2+ BC. Overexpression of GRB7 increased the proliferation, migration, invasion, and colony formation of HER2+ BC cells, while depletion of GRB7 had the opposite effects in HER2+ BC cells and inhibited xenograft growth. ChIP-PCR and luciferase reporter assay revealed that TCF12 directly bound to the promoter of the GRB7 gene to promote its transcription. GRB7 facilitated HER2+ BC epithelial-mesenchymal transition (EMT) progression by interacting with Notch1 to activate Wnt/β-catenin pathways and other signaling (i.e., AKT, ERK). Moreover, forced GRB7 overexpression activated Wnt/β-catenin to promote EMT progression, and partially rescued the inhibition of HER2+ BC proliferation, migration and invasion induced by TCF12 silencing. CONCLUSIONS Our work elucidates the oncogenic role of GRB7 in HER2+ BC, which could serve as a prognostic indicator and promising therapeutic target.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, China
| | - Yuanli Wu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, China
| | - Yue Su
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, China
| | - Na Qu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, China
| | - Bo Chen
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, China
| | - Duanfang Zhou
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, China
| | - Lie Yuan
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, China
| | - Manjialan Yin
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, China
| | - Mingpu Liu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, China
| | - Weiying Zhou
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China.
- Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China.
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
26
|
Zhu PK, Lin MX, Zeng MY, Tang Y, Li XR, He TY, Zheng YS, Chen LY. Expression of Iron Metabolism Genes Is Potentially Regulated by DOF Transcription Factors in Dendrocalamus latiflorus Leaves. Int J Mol Sci 2024; 25:8114. [PMID: 39125685 PMCID: PMC11311721 DOI: 10.3390/ijms25158114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Transcription factors (TFs) are crucial pre-transcriptional regulatory mechanisms that can modulate the expression of downstream genes by binding to their promoter regions. DOF (DNA binding with One Finger) proteins are a unique class of TFs with extensive roles in plant growth and development. Our previous research indicated that iron content varies among bamboo leaves of different colors. However, to our knowledge, genes related to iron metabolism pathways in bamboo species have not yet been studied. Therefore, in the current study, we identified iron metabolism related (IMR) genes in bamboo and determined the TFs that significantly influence them. Among these, DOFs were found to have widespread effects and potentially significant impacts on their expression. We identified specific DOF members in Dendrocalamus latiflorus with binding abilities through homology with Arabidopsis DOF proteins, and established connections between some of these members and IMR genes using RNA-seq data. Additionally, molecular docking confirmed the binding interactions between these DlDOFs and the DOF binding sites in the promoter regions of IMR genes. The co-expression relationship between the two gene sets was further validated using q-PCR experiments. This study paves the way for research into iron metabolism pathways in bamboo and lays the foundation for understanding the role of DOF TFs in D. latiflorus.
Collapse
Affiliation(s)
- Peng-Kai Zhu
- College of Landscape Architecture and Art, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Mei-Xia Lin
- College of Landscape Architecture and Art, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Mei-Yin Zeng
- College of Landscape Architecture and Art, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yu Tang
- College of Landscape Architecture and Art, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xin-Rui Li
- College of Landscape Architecture and Art, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Tian-You He
- College of Landscape Architecture and Art, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yu-Shan Zheng
- College of Landscape Architecture and Art, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- College of Forestry, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ling-Yan Chen
- College of Landscape Architecture and Art, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
27
|
Mayse L, Wang Y, Ahmad M, Movileanu L. Real-Time Measurement of a Weak Interaction of a Transcription Factor Motif with a Protein Hub at Single-Molecule Precision. ACS NANO 2024; 18:20468-20481. [PMID: 39049818 PMCID: PMC11308778 DOI: 10.1021/acsnano.4c04857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Transcription factors often interact with other protein cofactors, regulating gene expression. Direct detection of these brief events using existing technologies remains challenging due to their transient nature. In addition, intrinsically disordered domains, intranuclear location, and lack of cofactor-dependent active sites of transcription factors further complicate the quantitative analysis of these critical processes. Here, we create a genetically encoded label-free sensor to identify the interaction between a motif of the MYC transcription factor, a primary cancer driver, and WDR5, a chromatin-associated protein hub. Using an engineered nanopore equipped with this motif, WDR5 is probed through reversible captures and releases in a one-by-one and time-resolved fashion. Our single-molecule kinetic measurements indicate a weak-affinity interaction arising from a relatively slow complex association and a fast dissociation of WDR5 from the tethered motif. Further, we validate this subtle interaction by determinations in an ensemble using single nanodisc-wrapped nanopores immobilized on a biolayer interferometry sensor. This study also provides the proof-of-concept for a sensor that reveals unique recognition signatures of different protein binding sites. Our foundational work may be further developed to produce sensing elements for analytical proteomics and cancer nanomedicine.
Collapse
Affiliation(s)
- Lauren
A. Mayse
- Department
of Physics, Syracuse University, 201 Physics Building, Syracuse, New York 13244, United States
- Department
of Biomedical and Chemical Engineering, Syracuse University, 329 Link Hall, Syracuse, New York 13244, United States
| | - Yazheng Wang
- Department
of Physics, Syracuse University, 201 Physics Building, Syracuse, New York 13244, United States
- Department
of Biomedical and Chemical Engineering, Syracuse University, 329 Link Hall, Syracuse, New York 13244, United States
| | - Mohammad Ahmad
- Department
of Physics, Syracuse University, 201 Physics Building, Syracuse, New York 13244, United States
| | - Liviu Movileanu
- Department
of Physics, Syracuse University, 201 Physics Building, Syracuse, New York 13244, United States
- Department
of Biomedical and Chemical Engineering, Syracuse University, 329 Link Hall, Syracuse, New York 13244, United States
- Department
of Biology, Syracuse University, 114 Life Sciences Complex, Syracuse, New York 13244, United States
- The
BioInspired Institute, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
28
|
Munshi R. How Transcription Factor Clusters Shape the Transcriptional Landscape. Biomolecules 2024; 14:875. [PMID: 39062589 PMCID: PMC11274464 DOI: 10.3390/biom14070875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
In eukaryotic cells, gene transcription typically occurs in discrete periods of promoter activity, interspersed with intervals of inactivity. This pattern deviates from simple stochastic events and warrants a closer examination of the molecular interactions that activate the promoter. Recent studies have identified transcription factor (TF) clusters as key precursors to transcriptional bursting. Often, these TF clusters form at chromatin segments that are physically distant from the promoter, making changes in chromatin conformation crucial for promoter-TF cluster interactions. In this review, I explore the formation and constituents of TF clusters, examining how the dynamic interplay between chromatin architecture and TF clustering influences transcriptional bursting. Additionally, I discuss techniques for visualizing TF clusters and provide an outlook on understanding the remaining gaps in this field.
Collapse
Affiliation(s)
- Rahul Munshi
- Joseph Henry Laboratories of Physics and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
29
|
Murthy S, Dey U, Olymon K, Abbas E, Yella VR, Kumar A. Discerning the Role of DNA Sequence, Shape, and Flexibility in Recognition by Drosophila Transcription Factors. ACS Chem Biol 2024; 19:1533-1543. [PMID: 38902964 DOI: 10.1021/acschembio.4c00202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
The precise spatial and temporal orchestration of gene expression is crucial for the ontogeny of an organism and is mainly governed by transcription factors (TFs). The mechanism of recognition of cognate sites amid millions of base pairs in the genome by TFs is still incompletely understood. In this study, we focus on DNA sequence composition, shape, and flexibility preferences of 28 quintessential TFs from Drosophila melanogaster that are critical to development and body patterning mechanisms. Our study finds that TFs exhibit distinct predilections for DNA shape, flexibility, and sequence compositions in the proximity of transcription factor binding sites (TFBSs). Notably, certain zinc finger proteins prefer GC-rich areas with less negative propeller twist, while homeodomains mainly seek AT-rich regions with a more negative propeller twist at their sites. Intriguingly, while numerous cofactors share similar binding site preferences and bind closer to each other in the genome, some cofactors that have different preferences bind farther apart. These findings shed light on TF DNA recognition and provide novel insights into possible cofactor binding and transcriptional regulation mechanisms.
Collapse
Affiliation(s)
- Smrithi Murthy
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam 784028, India
| | - Upalabdha Dey
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam 784028, India
| | - Kaushika Olymon
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam 784028, India
| | - Eshan Abbas
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam 784028, India
| | - Venkata Rajesh Yella
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Guntur 520002, India
| | - Aditya Kumar
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam 784028, India
| |
Collapse
|
30
|
Hu S, Liu Y, Zhang Q, Bai J, Xu C. A continuum of zinc finger transcription factor retention on native chromatin underlies dynamic genome organization. Mol Syst Biol 2024; 20:799-824. [PMID: 38745107 PMCID: PMC11220090 DOI: 10.1038/s44320-024-00038-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Transcription factor (TF) residence on chromatin translates into quantitative transcriptional or structural outcomes on genome. Commonly used formaldehyde crosslinking fixes TF-DNA interactions cumulatively and compromises the measured occupancy level. Here we mapped the occupancy level of global or individual zinc finger TFs like CTCF and MAZ, in the form of highly resolved footprints, on native chromatin. By incorporating reinforcing perturbation conditions, we established S-score, a quantitative metric to proxy the continuum of CTCF or MAZ retention across different motifs on native chromatin. The native chromatin-retained CTCF sites harbor sequence features within CTCF motifs better explained by S-score than the metrics obtained from other crosslinking or native assays. CTCF retention on native chromatin correlates with local SUMOylation level, and anti-correlates with transcriptional activity. The S-score successfully delineates the otherwise-masked differential stability of chromatin structures mediated by CTCF, or by MAZ independent of CTCF. Overall, our study established a paradigm continuum of TF retention across binding sites on native chromatin, explaining the dynamic genome organization.
Collapse
Affiliation(s)
- Siling Hu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yangying Liu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qifan Zhang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Juan Bai
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chenhuan Xu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.
- China National Center for Bioinformation, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
31
|
Liu L, Yahaya BS, Li J, Wu F. Enigmatic role of auxin response factors in plant growth and stress tolerance. FRONTIERS IN PLANT SCIENCE 2024; 15:1398818. [PMID: 38903418 PMCID: PMC11188990 DOI: 10.3389/fpls.2024.1398818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024]
Abstract
Abiotic and biotic stresses globally constrain plant growth and impede the optimization of crop productivity. The phytohormone auxin is involved in nearly every aspect of plant development. Auxin acts as a chemical messenger that influences gene expression through a short nuclear pathway, mediated by a family of specific DNA-binding transcription factors known as Auxin Response Factors (ARFs). ARFs thus act as effectors of auxin response and translate chemical signals into the regulation of auxin responsive genes. Since the initial discovery of the first ARF in Arabidopsis, advancements in genetics, biochemistry, genomics, and structural biology have facilitated the development of models elucidating ARF action and their contributions to generating specific auxin responses. Yet, significant gaps persist in our understanding of ARF transcription factors despite these endeavors. Unraveling the functional roles of ARFs in regulating stress response, alongside elucidating their genetic and molecular mechanisms, is still in its nascent phase. Here, we review recent research outcomes on ARFs, detailing their involvement in regulating leaf, flower, and root organogenesis and development, as well as stress responses and their corresponding regulatory mechanisms: including gene expression patterns, functional characterization, transcriptional, post-transcriptional and post- translational regulation across diverse stress conditions. Furthermore, we delineate unresolved questions and forthcoming challenges in ARF research.
Collapse
Affiliation(s)
- Ling Liu
- Faculty of Agriculture, Forestry and Food Engineering, Yibin University, Yibin, Sichuan, China
| | - Baba Salifu Yahaya
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, Sichuan, China
- Key Laboratory of Biology and Genetic Improvement of Maize in Southwest Region, Ministry of Agriculture, Wenjiang, Sichuan, China
| | - Jing Li
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, Sichuan, China
- Key Laboratory of Biology and Genetic Improvement of Maize in Southwest Region, Ministry of Agriculture, Wenjiang, Sichuan, China
| | - Fengkai Wu
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, Sichuan, China
- Key Laboratory of Biology and Genetic Improvement of Maize in Southwest Region, Ministry of Agriculture, Wenjiang, Sichuan, China
| |
Collapse
|
32
|
Feng XA, Yamadi M, Fu Y, Ness KM, Liu C, Ahmed I, Bowman GD, Johnson ME, Ha T, Wu C. GAGA zinc finger transcription factor searches chromatin by 1D-3D facilitated diffusion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.14.549009. [PMID: 37502885 PMCID: PMC10369947 DOI: 10.1101/2023.07.14.549009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
To elucidate how eukaryotic sequence-specific transcription factors (TFs) search for gene targets on chromatin, we used multi-color smFRET and single-particle imaging to track the diffusion of purified GAGA-Associated Factor (GAF) on DNA and nucleosomes. Monomeric GAF DNA-binding domain (DBD) bearing one zinc finger finds its cognate site by 1D or 3D diffusion on bare DNA and rapidly slides back-and-forth between naturally clustered motifs for seconds before escape. Multimeric, full-length GAF also finds clustered motifs on DNA by 1D-3D diffusion, but remains locked on target for longer periods. Nucleosome architecture effectively blocks GAF-DBD 1D-sliding into the histone core but favors retention of GAF-DBD when targeting solvent-exposed sites by 3D-diffusion. Despite the occlusive power of nucleosomes, 1D-3D facilitated diffusion enables GAF to effectively search for clustered cognate motifs in chromatin, providing a mechanism for navigation to nucleosome and nucleosome-free sites by a member of the largest TF family.
Collapse
Affiliation(s)
- Xinyu A. Feng
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Maryam Yamadi
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yiben Fu
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kaitlin M. Ness
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Celina Liu
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ishtiyaq Ahmed
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gregory D. Bowman
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Margaret E. Johnson
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Taekjip Ha
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Boston, Massachusetts, USA
| | - Carl Wu
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
33
|
Gupta S, Kesarwani V, Bhati U, Jyoti, Shankar R. PTFSpot: deep co-learning on transcription factors and their binding regions attains impeccable universality in plants. Brief Bioinform 2024; 25:bbae324. [PMID: 39013383 PMCID: PMC11250369 DOI: 10.1093/bib/bbae324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/07/2024] [Accepted: 06/19/2024] [Indexed: 07/18/2024] Open
Abstract
Unlike animals, variability in transcription factors (TFs) and their binding regions (TFBRs) across the plants species is a major problem that most of the existing TFBR finding software fail to tackle, rendering them hardly of any use. This limitation has resulted into underdevelopment of plant regulatory research and rampant use of Arabidopsis-like model species, generating misleading results. Here, we report a revolutionary transformers-based deep-learning approach, PTFSpot, which learns from TF structures and their binding regions' co-variability to bring a universal TF-DNA interaction model to detect TFBR with complete freedom from TF and species-specific models' limitations. During a series of extensive benchmarking studies over multiple experimentally validated data, it not only outperformed the existing software by >30% lead but also delivered consistently >90% accuracy even for those species and TF families that were never encountered during the model-building process. PTFSpot makes it possible now to accurately annotate TFBRs across any plant genome even in the total lack of any TF information, completely free from the bottlenecks of species and TF-specific models.
Collapse
Affiliation(s)
- Sagar Gupta
- Studio of Computational Biology & Bioinformatics, The Himalayan Centre for High-throughput Computational Biology, (HiCHiCoB, A BIC supported by DBT, India), Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Veerbhan Kesarwani
- Studio of Computational Biology & Bioinformatics, The Himalayan Centre for High-throughput Computational Biology, (HiCHiCoB, A BIC supported by DBT, India), Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Umesh Bhati
- Studio of Computational Biology & Bioinformatics, The Himalayan Centre for High-throughput Computational Biology, (HiCHiCoB, A BIC supported by DBT, India), Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Jyoti
- Studio of Computational Biology & Bioinformatics, The Himalayan Centre for High-throughput Computational Biology, (HiCHiCoB, A BIC supported by DBT, India), Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Ravi Shankar
- Studio of Computational Biology & Bioinformatics, The Himalayan Centre for High-throughput Computational Biology, (HiCHiCoB, A BIC supported by DBT, India), Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
34
|
Tur S, Palii CG, Brand M. Cell fate decision in erythropoiesis: Insights from multiomics studies. Exp Hematol 2024; 131:104167. [PMID: 38262486 PMCID: PMC10939800 DOI: 10.1016/j.exphem.2024.104167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 01/25/2024]
Abstract
Every second, the body produces 2 million red blood cells through a process called erythropoiesis. Erythropoiesis is hierarchical in that it results from a series of cell fate decisions whereby hematopoietic stem cells progress toward the erythroid lineage. Single-cell transcriptomic and proteomic approaches have revolutionized the way we understand erythropoiesis, revealing it to be a gradual process that underlies a progressive restriction of fate potential driven by quantitative changes in lineage-specifying transcription factors. Despite these major advances, we still know very little about what cell fate decision entails at the molecular level. Novel approaches that simultaneously measure additional properties in single cells, including chromatin accessibility, transcription factor binding, and/or cell surface proteins are being developed at a fast pace, providing the means to exciting new advances in the near future. In this review, we briefly summarize the main findings obtained from single-cell studies of erythropoiesis, highlight outstanding questions, and suggest recent technological advances to address them.
Collapse
Affiliation(s)
- Steven Tur
- Department of Cell and Regenerative Biology, Wisconsin Blood Cancer Research Institute, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Carbone Cancer Center, Madison, WI; Cellular and Molecular Biology Graduate Program, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Carmen G Palii
- Department of Cell and Regenerative Biology, Wisconsin Blood Cancer Research Institute, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Carbone Cancer Center, Madison, WI
| | - Marjorie Brand
- Department of Cell and Regenerative Biology, Wisconsin Blood Cancer Research Institute, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Carbone Cancer Center, Madison, WI.
| |
Collapse
|
35
|
Das R, Sakaue T, Shivashankar GV, Prost J, Hiraiwa T. Chromatin Remodeling Due to Transient-Link-and-Pass Activity Enhances Subnuclear Dynamics. PHYSICAL REVIEW LETTERS 2024; 132:058401. [PMID: 38364140 DOI: 10.1103/physrevlett.132.058401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 01/02/2024] [Indexed: 02/18/2024]
Abstract
Spatiotemporal coordination of chromatin and subnuclear compartments is crucial for cells. Numerous enzymes act inside nucleus-some of those transiently link and pass two chromatin segments. Here, we study how such an active perturbation affects fluctuating dynamics of an inclusion in the chromatic medium. Using numerical simulations and a versatile effective model, we categorize inclusion dynamics into three distinct modes. The transient-link-and-pass activity speeds up inclusion dynamics by affecting a slow mode related to chromatin remodeling, viz., size and shape of the chromatin meshes.
Collapse
Affiliation(s)
- Rakesh Das
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Takahiro Sakaue
- Department of Physical Sciences, Aoyama Gakuin University, Kanagawa 252-5258, Japan
| | - G V Shivashankar
- Department of Health Sciences and Technology (D-HEST), ETH Zurich, Villigen 8092, Switzerland
- Division of Biology and Chemistry, Paul Scherrer Institute, Villigen 5232, Switzerland
| | - Jacques Prost
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Laboratoire Physico Chimie Curie, Institut Curie, Paris Science et Lettres Research University, 75005 Paris, France
| | - Tetsuya Hiraiwa
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Institute of Physics, Academia Sinica, Taipei City 115201, Taiwan
| |
Collapse
|
36
|
Zhang F, Zhou H, Xue J, Zhang Y, Zhou L, Leng J, Fang G, Liu Y, Wang Y, Liu H, Wu Y, Qi L, Duan R, He X, Wang Y, Liu Y, Li L, Yang J, Liang D, Chen YH. Deficiency of Transcription Factor Sp1 Contributes to Hypertrophic Cardiomyopathy. Circ Res 2024; 134:290-306. [PMID: 38197258 DOI: 10.1161/circresaha.123.323272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is the most prevalent monogenic heart disorder. However, the pathogenesis of HCM, especially its nongenetic mechanisms, remains largely unclear. Transcription factors are known to be involved in various biological processes including cell growth. We hypothesized that SP1 (specificity protein 1), the first purified TF in mammals, plays a role in the cardiomyocyte growth and cardiac hypertrophy of HCM. METHODS Cardiac-specific conditional knockout of Sp1 mice were constructed to investigate the role of SP1 in the heart. The echocardiography, histochemical experiment, and transmission electron microscope were performed to analyze the cardiac phenotypes of cardiac-specific conditional knockout of Sp1 mice. RNA sequencing, chromatin immunoprecipitation sequencing, and adeno-associated virus experiments in vivo were performed to explore the downstream molecules of SP1. To examine the therapeutic effect of SP1 on HCM, an SP1 overexpression vector was constructed and injected into the mutant allele of Myh6 R404Q/+ (Myh6 c. 1211C>T) HCM mice. The human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from a patient with HCM were used to detect the potential therapeutic effects of SP1 in human HCM. RESULTS The cardiac-specific conditional knockout of Sp1 mice developed a typical HCM phenotype, displaying overt myocardial hypertrophy, interstitial fibrosis, and disordered myofilament. In addition, Sp1 knockdown dramatically increased the cell area of hiPSC-CMs and caused intracellular myofibrillar disorganization, which was similar to the hypertrophic cardiomyocytes of HCM. Mechanistically, Tuft1 was identified as the key target gene of SP1. The hypertrophic phenotypes induced by Sp1 knockdown in both hiPSC-CMs and mice could be rescued by TUFT1 (tuftelin 1) overexpression. Furthermore, SP1 overexpression suppressed the development of HCM in the mutant allele of Myh6 R404Q/+ mice and also reversed the hypertrophic phenotype of HCM hiPSC-CMs. CONCLUSIONS Our study demonstrates that SP1 deficiency leads to HCM. SP1 overexpression exhibits significant therapeutic effects on both HCM mice and HCM hiPSC-CMs, suggesting that SP1 could be a potential intervention target for HCM.
Collapse
Affiliation(s)
- Fulei Zhang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Huixing Zhou
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Jinfeng Xue
- Department of Regenerative Medicine (J.X., L.Q.), Tongji University School of Medicine, Shanghai, China
| | - Yuemei Zhang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Liping Zhou
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Junwei Leng
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Guojian Fang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yuanyuan Liu
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Jinzhou Medical University, China (Yuanyuan Liu, Y. Wang, Yan Wang)
| | - Yan Wang
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Jinzhou Medical University, China (Yuanyuan Liu, Y. Wang, Yan Wang)
| | - Hongyu Liu
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yahan Wu
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Lingbin Qi
- Department of Regenerative Medicine (J.X., L.Q.), Tongji University School of Medicine, Shanghai, China
| | - Ran Duan
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Xiaoyu He
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yan Wang
- Jinzhou Medical University, China (Yuanyuan Liu, Y. Wang, Yan Wang)
| | - Yi Liu
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Li Li
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Pathology and Pathophysiology (L.L., J.Y., Y.-H.C.), Tongji University School of Medicine, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China (L.L., J.Y., D.L., Y.-H.C.)
| | - Jian Yang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Pathology and Pathophysiology (L.L., J.Y., Y.-H.C.), Tongji University School of Medicine, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China (L.L., J.Y., D.L., Y.-H.C.)
| | - Dandan Liang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China (L.L., J.Y., D.L., Y.-H.C.)
| | - Yi-Han Chen
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Pathology and Pathophysiology (L.L., J.Y., Y.-H.C.), Tongji University School of Medicine, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China (L.L., J.Y., D.L., Y.-H.C.)
| |
Collapse
|
37
|
Khanbabei A, Segura L, Petrossian C, Lemus A, Cano I, Frazier C, Halajyan A, Ca D, Loza-Coll M. Experimental validation and characterization of putative targets of Escargot and STAT, two master regulators of the intestinal stem cells in Drosophila melanogaster. Dev Biol 2024; 505:148-163. [PMID: 37952851 DOI: 10.1016/j.ydbio.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 10/15/2023] [Accepted: 10/25/2023] [Indexed: 11/14/2023]
Abstract
Many organs contain adult stem cells (ASCs) to replace cells due to damage, disease, or normal tissue turnover. ASCs can divide asymmetrically, giving rise to a new copy of themselves (self-renewal) and a sister that commits to a specific cell type (differentiation). Decades of research have led to the identification of pleiotropic genes whose loss or gain of function affect diverse aspects of normal ASC biology. Genome-wide screens of these so-called genetic "master regulator" (MR) genes, have pointed to hundreds of putative targets that could serve as their downstream effectors. Here, we experimentally validate and characterize the regulation of several putative targets of Escargot (Esg) and the Signal Transducer and Activator of Transcription (Stat92E, a.k.a. STAT), two known MRs in Drosophila intestinal stem cells (ISCs). Our results indicate that regardless of bioinformatic predictions, most experimentally validated targets show a profile of gene expression that is consistent with co-regulation by both Esg and STAT, fitting a rather limited set of co-regulatory modalities. A bioinformatic analysis of proximal regulatory sequences in specific subsets of co-regulated targets identified additional transcription factors that might cooperate with Esg and STAT in modulating their transcription. Lastly, in vivo manipulations of validated targets rarely phenocopied the effects of manipulating Esg and STAT, suggesting the existence of complex genetic interactions among downstream targets of these two MR genes during ISC homeostasis.
Collapse
Affiliation(s)
- Armen Khanbabei
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Lina Segura
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Cynthia Petrossian
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Aaron Lemus
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Ithan Cano
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Courtney Frazier
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Armen Halajyan
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Donnie Ca
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Mariano Loza-Coll
- Department of Biology, California State University, Northridge (CSUN), USA.
| |
Collapse
|
38
|
Nagpal H, Ali-Ahmad A, Hirano Y, Cai W, Halic M, Fukagawa T, Sekulić N, Fierz B. CENP-A and CENP-B collaborate to create an open centromeric chromatin state. Nat Commun 2023; 14:8227. [PMID: 38086807 PMCID: PMC10716449 DOI: 10.1038/s41467-023-43739-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Centromeres are epigenetically defined via the presence of the histone H3 variant CENP-A. Contacting CENP-A nucleosomes, the constitutive centromere associated network (CCAN) and the kinetochore assemble, connecting the centromere to spindle microtubules during cell division. The DNA-binding centromeric protein CENP-B is involved in maintaining centromere stability and, together with CENP-A, shapes the centromeric chromatin state. The nanoscale organization of centromeric chromatin is not well understood. Here, we use single-molecule fluorescence and cryoelectron microscopy (cryoEM) to show that CENP-A incorporation establishes a dynamic and open chromatin state. The increased dynamics of CENP-A chromatin create an opening for CENP-B DNA access. In turn, bound CENP-B further opens the chromatin fiber structure and induces nucleosomal DNA unwrapping. Finally, removal of CENP-A increases CENP-B mobility in cells. Together, our studies show that the two centromere-specific proteins collaborate to reshape chromatin structure, enabling the binding of centromeric factors and establishing a centromeric chromatin state.
Collapse
Affiliation(s)
- Harsh Nagpal
- École Polytechnique Fédérale de Lausanne (EPFL), SB ISIC LCBM, Station 6, CH-1015, Lausanne, Switzerland
| | - Ahmad Ali-Ahmad
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, Faculty of Medicine, University of Oslo, Oslo, 0318, Norway
| | - Yasuhiro Hirano
- Graduate School of Frontier Biosciences, Osaka University, Suita, 565-0871, Japan
| | - Wei Cai
- École Polytechnique Fédérale de Lausanne (EPFL), SB ISIC LCBM, Station 6, CH-1015, Lausanne, Switzerland
| | - Mario Halic
- Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105-3678, USA
| | - Tatsuo Fukagawa
- Graduate School of Frontier Biosciences, Osaka University, Suita, 565-0871, Japan
| | - Nikolina Sekulić
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, Faculty of Medicine, University of Oslo, Oslo, 0318, Norway.
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, 0315, Norway.
| | - Beat Fierz
- École Polytechnique Fédérale de Lausanne (EPFL), SB ISIC LCBM, Station 6, CH-1015, Lausanne, Switzerland.
| |
Collapse
|
39
|
Talukdar PD, Chatterji U. Transcriptional co-activators: emerging roles in signaling pathways and potential therapeutic targets for diseases. Signal Transduct Target Ther 2023; 8:427. [PMID: 37953273 PMCID: PMC10641101 DOI: 10.1038/s41392-023-01651-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/27/2023] [Accepted: 09/10/2023] [Indexed: 11/14/2023] Open
Abstract
Specific cell states in metazoans are established by the symphony of gene expression programs that necessitate intricate synergic interactions between transcription factors and the co-activators. Deregulation of these regulatory molecules is associated with cell state transitions, which in turn is accountable for diverse maladies, including developmental disorders, metabolic disorders, and most significantly, cancer. A decade back most transcription factors, the key enablers of disease development, were historically viewed as 'undruggable'; however, in the intervening years, a wealth of literature validated that they can be targeted indirectly through transcriptional co-activators, their confederates in various physiological and molecular processes. These co-activators, along with transcription factors, have the ability to initiate and modulate transcription of diverse genes necessary for normal physiological functions, whereby, deregulation of such interactions may foster tissue-specific disease phenotype. Hence, it is essential to analyze how these co-activators modulate specific multilateral processes in coordination with other factors. The proposed review attempts to elaborate an in-depth account of the transcription co-activators, their involvement in transcription regulation, and context-specific contributions to pathophysiological conditions. This review also addresses an issue that has not been dealt with in a comprehensive manner and hopes to direct attention towards future research that will encompass patient-friendly therapeutic strategies, where drugs targeting co-activators will have enhanced benefits and reduced side effects. Additional insights into currently available therapeutic interventions and the associated constraints will eventually reveal multitudes of advanced therapeutic targets aiming for disease amelioration and good patient prognosis.
Collapse
Affiliation(s)
- Priyanka Dey Talukdar
- Cancer Research Laboratory, Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, West Bengal, India
| | - Urmi Chatterji
- Cancer Research Laboratory, Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, West Bengal, India.
| |
Collapse
|
40
|
Höpfner D, Cichy A, Pogenberg V, Krisp C, Mezouar S, Bach NC, Grotheer J, Zarza SM, Martinez E, Bonazzi M, Feige MJ, Sieber SA, Schlüter H, Itzen A. The DNA-binding induced (de)AMPylation activity of a Coxiella burnetii Fic enzyme targets Histone H3. Commun Biol 2023; 6:1124. [PMID: 37932372 PMCID: PMC10628234 DOI: 10.1038/s42003-023-05494-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 10/20/2023] [Indexed: 11/08/2023] Open
Abstract
The intracellular bacterial pathogen Coxiella burnetii evades the host response by secreting effector proteins that aid in establishing a replication-friendly niche. Bacterial filamentation induced by cyclic AMP (Fic) enzymes can act as effectors by covalently modifying target proteins with the posttranslational AMPylation by transferring adenosine monophosphate (AMP) from adenosine triphosphate (ATP) to a hydroxyl-containing side chain. Here we identify the gene product of C. burnetii CBU_0822, termed C. burnetii Fic 2 (CbFic2), to AMPylate host cell histone H3 at serine 10 and serine 28. We show that CbFic2 acts as a bifunctional enzyme, both capable of AMPylation as well as deAMPylation, and is regulated by the binding of DNA via a C-terminal helix-turn-helix domain. We propose that CbFic2 performs AMPylation in its monomeric state, switching to a deAMPylating dimer upon DNA binding. This study unveils reversible histone modification by a specific enzyme of a pathogenic bacterium.
Collapse
Affiliation(s)
- Dorothea Höpfner
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246, Hamburg, Germany
| | - Adam Cichy
- Center for Integrated Protein Science Munich (CIPSM), Department Chemistry, Group of Proteinchemistry, Technical University of Munich, Lichtenbergstraße 4, 85747, Garching, Germany
| | - Vivian Pogenberg
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246, Hamburg, Germany
| | - Christoph Krisp
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246, Hamburg, Germany
| | - Soraya Mezouar
- Aix-Marseille University, Institut de Recherche pour la Développement (IRD), Assistance Publique-Hôpitaux de Marseille (APHM), Microbes Evolution Phylogeny and Infections (MEPHI), Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Boulevard Jean Moulin, 13005, Marseille, France
| | - Nina C Bach
- Technical University of Munich (TUM), TUM School of Natural Sciences, Department of Biosciences, Chair of Organic Chemistry II, Center for Functional Protein Assemblies (CPA), Ernst-Otto-Fischer Straße 8, 85748, Garching, Germany
| | - Jan Grotheer
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246, Hamburg, Germany
| | - Sandra Madariaga Zarza
- Aix-Marseille University, Institut de Recherche pour la Développement (IRD), Assistance Publique-Hôpitaux de Marseille (APHM), Microbes Evolution Phylogeny and Infections (MEPHI), Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Boulevard Jean Moulin, 13005, Marseille, France
| | - Eric Martinez
- Cellular and Molecular Biology of Bacterial Infections, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, UMR 9004 - Centre national de la recherche scientifique (CNRS), 1919 Route de Mende, 34293, Montpellier, France
| | - Matteo Bonazzi
- Cellular and Molecular Biology of Bacterial Infections, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, UMR 9004 - Centre national de la recherche scientifique (CNRS), 1919 Route de Mende, 34293, Montpellier, France
| | - Matthias J Feige
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Lichtenbergstraße 4, 85748, Garching, Germany
| | - Stephan A Sieber
- Technical University of Munich (TUM), TUM School of Natural Sciences, Department of Biosciences, Chair of Organic Chemistry II, Center for Functional Protein Assemblies (CPA), Ernst-Otto-Fischer Straße 8, 85748, Garching, Germany
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246, Hamburg, Germany
| | - Aymelt Itzen
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246, Hamburg, Germany.
- Center for Structural Systems Biology (CSSB), University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
41
|
Wu R, Zhou B, Wang W, Liu F. Regulatory Mechanisms for Transcriptional Bursting Revealed by an Event-Based Model. RESEARCH (WASHINGTON, D.C.) 2023; 6:0253. [PMID: 39290237 PMCID: PMC11407585 DOI: 10.34133/research.0253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/01/2023] [Indexed: 09/19/2024]
Abstract
Gene transcription often occurs in discrete bursts, and it can be difficult to deduce the underlying regulatory mechanisms for transcriptional bursting with limited experimental data. Here, we categorize numerous states of single eukaryotic genes and identify 6 essential transcriptional events, each comprising a series of state transitions; transcriptional bursting is characterized as a sequence of 4 events, capable of being organized in various configurations, in addition to the beginning and ending events. By associating transcriptional kinetics with mean durations and recurrence probabilities of the events, we unravel how transcriptional bursting is modulated by various regulators including transcription factors. Through analytical derivation and numerical simulation, this study reveals key state transitions contributing to transcriptional sensitivity and specificity, typical characteristics of burst profiles, global constraints on intrinsic transcriptional noise, major regulatory modes in individual genes and across the genome, and requirements for fast gene induction upon stimulation. It is illustrated how biochemical reactions on different time scales are modulated to separately shape the durations and ordering of the events. Our results suggest that transcriptional patterns are essentially controlled by a shared set of transcriptional events occurring under specific promoter architectures and regulatory modes, the number of which is actually limited.
Collapse
Affiliation(s)
- Renjie Wu
- National Laboratory of Solid State Microstructures, Department of Physics, and Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, P. R. China
| | - Bangyan Zhou
- National Laboratory of Solid State Microstructures, Department of Physics, and Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, P. R. China
| | - Wei Wang
- National Laboratory of Solid State Microstructures, Department of Physics, and Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, P. R. China
- Institute for Brain Sciences, Nanjing University, Nanjing 210093, P. R. China
| | - Feng Liu
- National Laboratory of Solid State Microstructures, Department of Physics, and Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, P. R. China
- Institute for Brain Sciences, Nanjing University, Nanjing 210093, P. R. China
| |
Collapse
|
42
|
Zhu PK, Yang J, Yang DM, Xu YP, He TY, Rong JD, Zheng YS, Chen LY. Identification and characterization of the cupin_1 domain-containing proteins in ma bamboo ( Dendrocalamus latiflorus) and their potential role in rhizome sprouting. FRONTIERS IN PLANT SCIENCE 2023; 14:1260856. [PMID: 37908839 PMCID: PMC10614299 DOI: 10.3389/fpls.2023.1260856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/25/2023] [Indexed: 11/02/2023]
Abstract
Cupin_1 domain-containing protein (CDP) family, which is a member of the cupin superfamily with the most diverse functions in plants, has been found to be involved in hormone pathways that are closely related to rhizome sprouting (RS), a vital form of asexual reproduction in plants. Ma bamboo is a typical clumping bamboo, which mainly reproduces by RS. In this study, we identified and characterized 53 Dendrocalamus latiflorus CDP genes and divided them into seven subfamilies. Comparing the genetic structures among subfamilies showed a relatively conserved gene structure within each subfamily, and the number of cupin_1 domains affected the conservation among D. latiflorus CDP genes. Gene collinearity results showed that segmental duplication and tandem duplication both contributed to the expansion of D. latiflorus CDP genes, and lineage-specific gene duplication was an important factor influencing the evolution of CDP genes. Expression patterns showed that CDP genes generally had higher expression levels in germinating underground buds, indicating that they might play important roles in promoting shoot sprouting. Transcription factor binding site prediction and co-expression network analysis indicated that D. latiflorus CDPs were regulated by a large number of transcription factors, and collectively participated in rhizome buds and shoot development. This study significantly provided new insights into the evolutionary patterns and molecular functions of CDP genes, and laid a foundation for further studying the regulatory mechanisms of plant rhizome sprouting.
Collapse
Affiliation(s)
- Peng-kai Zhu
- College of Landscape Architecture and Art, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jing Yang
- College of Forestry, Fujian Agriculture and Forestry University, Fuzhou, China
| | - De-ming Yang
- College of Forestry, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yan-ping Xu
- College of Forestry, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Tian-you He
- College of Landscape Architecture and Art, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jun-dong Rong
- College of Forestry, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yu-shan Zheng
- College of Forestry, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ling-yan Chen
- College of Landscape Architecture and Art, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
43
|
Abstract
Cells must tightly regulate their gene expression programs and yet rapidly respond to acute biochemical and biophysical cues within their environment. This information is transmitted to the nucleus through various signaling cascades, culminating in the activation or repression of target genes. Transcription factors (TFs) are key mediators of these signals, binding to specific regulatory elements within chromatin. While live-cell imaging has conclusively proven that TF-chromatin interactions are highly dynamic, how such transient interactions can have long-term impacts on developmental trajectories and disease progression is still largely unclear. In this review, we summarize our current understanding of the dynamic nature of TF functions, starting with a historical overview of early live-cell experiments. We highlight key factors that govern TF dynamics and how TF dynamics, in turn, affect downstream transcriptional bursting. Finally, we conclude with open challenges and emerging technologies that will further our understanding of transcriptional regulation.
Collapse
Affiliation(s)
- Kaustubh Wagh
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; , ,
- Department of Physics, University of Maryland, College Park, Maryland, USA;
| | - Diana A Stavreva
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; , ,
| | - Arpita Upadhyaya
- Department of Physics, University of Maryland, College Park, Maryland, USA;
- Institute for Physical Science and Technology, University of Maryland, College Park, Maryland, USA
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; , ,
| |
Collapse
|
44
|
Mazzocca M, Loffreda A, Colombo E, Fillot T, Gnani D, Falletta P, Monteleone E, Capozi S, Bertrand E, Legube G, Lavagnino Z, Tacchetti C, Mazza D. Chromatin organization drives the search mechanism of nuclear factors. Nat Commun 2023; 14:6433. [PMID: 37833263 PMCID: PMC10575952 DOI: 10.1038/s41467-023-42133-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
Nuclear factors rapidly scan the genome for their targets, but the role of nuclear organization in such search is uncharted. Here we analyzed how multiple factors explore chromatin, combining live-cell single-molecule tracking with multifocal structured illumination of DNA density. We find that factors displaying higher bound fractions sample DNA-dense regions more exhaustively. Focusing on the tumor-suppressor p53, we demonstrate that it searches for targets by alternating between rapid diffusion in the interchromatin compartment and compact sampling of chromatin dense regions. Efficient targeting requires balanced interactions with chromatin: fusing p53 with an exogenous intrinsically disordered region potentiates p53-mediated target gene activation at low concentrations, but leads to condensates at higher levels, derailing its search and downregulating transcription. Our findings highlight the role of disordered regions on factors search and showcase a powerful method to generate traffic maps of the eukaryotic nucleus to dissect how its organization guides nuclear factors action.
Collapse
Affiliation(s)
- Matteo Mazzocca
- Università Vita-Salute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Alessia Loffreda
- IRCCS Ospedale San Raffaele, Experimental Imaging Center, Via Olgettina 58, 20132, Milan, Italy
| | - Emanuele Colombo
- Università Vita-Salute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Tom Fillot
- Università Vita-Salute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
- IRCCS Ospedale San Raffaele, Experimental Imaging Center, Via Olgettina 58, 20132, Milan, Italy
| | - Daniela Gnani
- Università Vita-Salute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Paola Falletta
- Università Vita-Salute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | | | - Serena Capozi
- Institut de Génétique Moléculaire de Montpellier, CNRS, Montpellier, 34293, France
| | - Edouard Bertrand
- Institut de Génétique Moléculaire de Montpellier, CNRS, Montpellier, 34293, France
| | - Gaelle Legube
- MCD, Centre de Biologie Intégrative (CBI), CNRS, Université de Toulouse, UT3, Toulouse, France
| | - Zeno Lavagnino
- IRCCS Ospedale San Raffaele, Experimental Imaging Center, Via Olgettina 58, 20132, Milan, Italy
- IFOM ETS- The AIRC Institute of Molecular Oncology-Via Adamello 16, 20139, Milan, Italy
| | - Carlo Tacchetti
- Università Vita-Salute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
- IRCCS Ospedale San Raffaele, Experimental Imaging Center, Via Olgettina 58, 20132, Milan, Italy
| | - Davide Mazza
- Università Vita-Salute San Raffaele, Via Olgettina 58, 20132, Milan, Italy.
- IRCCS Ospedale San Raffaele, Experimental Imaging Center, Via Olgettina 58, 20132, Milan, Italy.
| |
Collapse
|
45
|
Samee MAH. Noncanonical binding of transcription factors: time to revisit specificity? Mol Biol Cell 2023; 34:pe4. [PMID: 37486893 PMCID: PMC10398899 DOI: 10.1091/mbc.e22-08-0325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 06/05/2023] [Accepted: 06/21/2023] [Indexed: 07/26/2023] Open
Abstract
Transcription factors (TFs) are one of the most studied classes of DNA-binding proteins that have a direct functional impact on gene transcription and thus, on human physiology and disease. The mechanisms that TFs use for recognizing target DNA binding sites have been studied for nearly five decades, yet they remain poorly understood. It is classically assumed that a TF recognizes a specific sequence pattern, or motif, as its binding sites. However, recent studies are consistently finding examples of noncanonical binding, that is, TFs binding at sites that do not resemble their sequence motifs. Here we review the current literature on four major types of noncanonical TF binding, namely binding based on DNA shape readout, at Guanine-quadruplex structures, at repeat sequences, and bispecific binding. These examples point to a critical need for studies to unify our current observations, many of which are at odds with the "one TF, one motif" view, into a more comprehensive definition of the DNA-binding specificity of TFs.
Collapse
|
46
|
Lerner J, Katznelson A, Zhang J, Zaret KS. Different chromatin-scanning modes lead to targeting of compacted chromatin by pioneer factors FOXA1 and SOX2. Cell Rep 2023; 42:112748. [PMID: 37405916 PMCID: PMC10529229 DOI: 10.1016/j.celrep.2023.112748] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/20/2023] [Accepted: 06/19/2023] [Indexed: 07/07/2023] Open
Abstract
Pioneer transcription factors interact with nucleosomes to scan silent, compact chromatin, enabling cooperative events that modulate gene activity. While at a subset of sites pioneer factors access chromatin by assisted loading with other transcription factors, the nucleosome-binding properties of pioneer factors enable them to initiate zygotic genome activation, embryonic development, and cellular reprogramming. To better understand nucleosome targeting in vivo, we assess whether pioneer factors FoxA1 and Sox2 target stable or unstable nucleosomes and find that they target DNase-resistant, stable nucleosomes, whereas HNF4A, a non-nucleosome binding factor, targets open, DNase-sensitive chromatin. Despite FOXA1 and SOX2 targeting similar proportions of DNase-resistant chromatin, using single-molecule tracking, we find that FOXA1 uses lower nucleoplasmic diffusion and longer residence times while SOX2 uses higher nucleoplasmic diffusion and shorter residence times to scan compact chromatin, while HNF4 scans compact chromatin much less efficiently. Thus, pioneer factors target compact chromatin through distinct processes.
Collapse
Affiliation(s)
- Jonathan Lerner
- Institute for Regenerative Medicine and Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6058, USA
| | - Andrew Katznelson
- Institute for Regenerative Medicine and Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6058, USA
| | - Jingchao Zhang
- Institute for Regenerative Medicine and Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6058, USA
| | - Kenneth S Zaret
- Institute for Regenerative Medicine and Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6058, USA.
| |
Collapse
|
47
|
Boumpas P, Merabet S, Carnesecchi J. Integrating transcription and splicing into cell fate: Transcription factors on the block. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1752. [PMID: 35899407 DOI: 10.1002/wrna.1752] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/22/2022] [Accepted: 07/01/2022] [Indexed: 11/10/2022]
Abstract
Transcription factors (TFs) are present in all life forms and conserved across great evolutionary distances in eukaryotes. From yeast to complex multicellular organisms, they are pivotal players of cell fate decision by orchestrating gene expression at diverse molecular layers. Notably, TFs fine-tune gene expression by coordinating RNA fate at both the expression and splicing levels. They regulate alternative splicing, an essential mechanism for cell plasticity, allowing the production of many mRNA and protein isoforms in precise cell and tissue contexts. Despite this apparent role in splicing, how TFs integrate transcription and splicing to ultimately orchestrate diverse cell functions and cell fate decisions remains puzzling. We depict substantial studies in various model organisms underlining the key role of TFs in alternative splicing for promoting tissue-specific functions and cell fate. Furthermore, we emphasize recent advances describing the molecular link between the transcriptional and splicing activities of TFs. As TFs can bind both DNA and/or RNA to regulate transcription and splicing, we further discuss their flexibility and compatibility for DNA and RNA substrates. Finally, we propose several models integrating transcription and splicing activities of TFs in the coordination and diversification of cell and tissue identities. This article is categorized under: RNA Processing > Splicing Regulation/Alternative Splicing RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications RNA Processing > Splicing Mechanisms.
Collapse
Affiliation(s)
- Panagiotis Boumpas
- Institut de Génomique Fonctionnelle de Lyon, UMR5242, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, Université Claude Bernard-Lyon 1, Lyon, France
| | - Samir Merabet
- Institut de Génomique Fonctionnelle de Lyon, UMR5242, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, Université Claude Bernard-Lyon 1, Lyon, France
| | - Julie Carnesecchi
- Institut de Génomique Fonctionnelle de Lyon, UMR5242, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, Université Claude Bernard-Lyon 1, Lyon, France
| |
Collapse
|
48
|
Roura AJ, Szadkowska P, Poleszak K, Dabrowski MJ, Ellert-Miklaszewska A, Wojnicki K, Ciechomska IA, Stepniak K, Kaminska B, Wojtas B. Regulatory networks driving expression of genes critical for glioblastoma are controlled by the transcription factor c-Jun and the pre-existing epigenetic modifications. Clin Epigenetics 2023; 15:29. [PMID: 36850002 PMCID: PMC9972689 DOI: 10.1186/s13148-023-01446-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM, WHO grade IV) is an aggressive, primary brain tumor. Despite extensive tumor resection followed by radio- and chemotherapy, life expectancy of GBM patients did not improve over decades. Several studies reported transcription deregulation in GBMs, but regulatory mechanisms driving overexpression of GBM-specific genes remain largely unknown. Transcription in open chromatin regions is directed by transcription factors (TFs) that bind to specific motifs, recruit co-activators/repressors and the transcriptional machinery. Identification of GBM-related TFs-gene regulatory networks may reveal new and targetable mechanisms of gliomagenesis. RESULTS We predicted TFs-regulated networks in GBMs in silico and intersected them with putative TF binding sites identified in the accessible chromatin in human glioma cells and GBM patient samples. The Cancer Genome Atlas and Glioma Atlas datasets (DNA methylation, H3K27 acetylation, transcriptomic profiles) were explored to elucidate TFs-gene regulatory networks and effects of the epigenetic background. In contrast to the majority of tumors, c-Jun expression was higher in GBMs than in normal brain and c-Jun binding sites were found in multiple genes overexpressed in GBMs, including VIM, FOSL2 or UPP1. Binding of c-Jun to the VIM gene promoter was stronger in GBM-derived cells than in cells derived from benign glioma as evidenced by gel shift and supershift assays. Regulatory regions of the majority of c-Jun targets have distinct DNA methylation patterns in GBMs as compared to benign gliomas, suggesting the contribution of DNA methylation to the c-Jun-dependent gene expression. CONCLUSIONS GBM-specific TFs-gene networks identified in GBMs differ from regulatory pathways attributed to benign brain tumors and imply a decisive role of c-Jun in controlling genes that drive glioma growth and invasion as well as a modulatory role of DNA methylation.
Collapse
Affiliation(s)
- Adria-Jaume Roura
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Paulina Szadkowska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Poleszak
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Michal J. Dabrowski
- Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | | | - Kamil Wojnicki
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Iwona A. Ciechomska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Karolina Stepniak
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bartosz Wojtas
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
- Laboratory of Sequencing, Nencki Institute of Experimental Biology, ul. Ludwika Pasteura 3, 02-093 Warsaw, Poland
| |
Collapse
|
49
|
Park JW, Lee EJ, Moon E, Kim HL, Kim IB, Hodzic D, Kim N, Kweon HS, Kim JW. Orthodenticle homeobox 2 is transported to lysosomes by nuclear budding vesicles. Nat Commun 2023; 14:1111. [PMID: 36849521 PMCID: PMC9971051 DOI: 10.1038/s41467-023-36697-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 02/08/2023] [Indexed: 03/01/2023] Open
Abstract
Transcription factors (TFs) are transported from the cytoplasm to the nucleus and disappear from the nucleus after they regulate gene expression. Here, we discover an unconventional nuclear export of the TF, orthodenticle homeobox 2 (OTX2), in nuclear budding vesicles, which transport OTX2 to the lysosome. We further find that torsin1a (Tor1a) is responsible for scission of the inner nuclear vesicle, which captures OTX2 using the LINC complex. Consistent with this, in cells expressing an ATPase-inactive Tor1aΔE mutant and the LINC (linker of nucleoskeleton and cytoskeleton) breaker KASH2, OTX2 accumulated and formed aggregates in the nucleus. Consequently, in the mice expressing Tor1aΔE and KASH2, OTX2 could not be secreted from the choroid plexus for transfer to the visual cortex, leading to failed development of parvalbumin neurons and reduced visual acuity. Together, our results suggest that unconventional nuclear egress and secretion of OTX2 are necessary not only to induce functional changes in recipient cells but also to prevent aggregation in donor cells.
Collapse
Affiliation(s)
- Jun Woo Park
- Department of Biological Sciences and Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea
| | - Eun Jung Lee
- Department of Biological Sciences and Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea
| | - Eunyoung Moon
- Electron Microscopy Research Center, Korea Basic Science Institute, Cheongju, 28119, South Korea
| | - Hong-Lim Kim
- Integrative Research Support Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, South Korea
| | - In-Beom Kim
- Integrative Research Support Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, South Korea
| | - Didier Hodzic
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Namsuk Kim
- Department of Biological Sciences and Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea.,Neurovascular Unit, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Hee-Seok Kweon
- Electron Microscopy Research Center, Korea Basic Science Institute, Cheongju, 28119, South Korea
| | - Jin Woo Kim
- Department of Biological Sciences and Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea.
| |
Collapse
|
50
|
Single cell epigenomic and transcriptomic analysis uncovers potential transcription factors regulating mitotic/meiotic switch. Cell Death Dis 2023; 14:134. [PMID: 36797258 PMCID: PMC9935506 DOI: 10.1038/s41419-023-05671-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/18/2023]
Abstract
In order to reveal the complex mechanism governing the mitotic/meiotic switch in female germ cells at epigenomic and genomic levels, we examined the chromatin accessibility (scATAC-seq) and the transcriptional dynamics (scRNA-seq) in germ cells of mouse embryonic ovary between E11.5 to 13.5 at single-cell resolution. Adopting a strict transcription factors (TFs) screening framework that makes it easier to understand the single-cell chromatin signature and a TF interaction algorithm that integrates the transcript levels, chromatin accessibility, and motif scores, we identified 14 TFs potentially regulating the mitotic/meiotic switch, including TCFL5, E2F1, E2F2, E2F6, E2F8, BATF3, SP1, FOS, FOXN3, VEZF1, GBX2, CEBPG, JUND, and TFDP1. Focusing on TCFL5, we constructed Tcfl5+/- mice which showed significantly reduced fertility and found that decreasing TCFL5 expression in cultured E12.5 ovaries by RNAi impaired meiotic progression from leptotene to zygotene. Bioinformatics analysis of published results of the embryonic germ cell transcriptome and the finding that in these cells central meiotic genes (Stra8, Tcfl5, Sycp3, and E2f2) possess open chromatin status already at the mitotic stage together with other features of TCFL5 (potential capability to interact with core TFs and activate meiotic genes, its progressive activation after preleptotene, binding sites in the promoter region of E2f2 and Sycp3), indicated extensive amplification of transcriptional programs associated to mitotic/meiotic switch with an important contribution of TCFL5. We conclude that the identified TFs, are involved in various stages of the mitotic/meiotic switch in female germ cells, TCFL5 primarily in meiotic progression. Further investigation on these factors might give a significant contribution to unravel the molecular mechanisms of this fundamental process of oogenesis and provide clues about pathologies in women such as primary ovarian insufficiency (POI) due at least in part to meiotic defects.
Collapse
|