1
|
Tan L, Xie XS, Lomvardas S. Genomic snowflakes: how the uniqueness of DNA folding allows us to smell the chemical universe. Curr Opin Genet Dev 2025; 92:102329. [PMID: 40107115 PMCID: PMC12068986 DOI: 10.1016/j.gde.2025.102329] [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: 12/16/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Olfactory receptor (OR) gene choice, the stable expression of one out of >2000 OR alleles by olfactory sensory neurons, constitutes a gene regulatory process that is driven by three-dimensional nuclear architecture. Moreover, the differentiation-dependent process that culminates in monogenic and monoallelic OR transcription represents a powerful demonstration of the rich mechanistic insight that single-cell genomics and multiomics can provide toward the understanding of a biological process. At this review, we describe the latest advances in the understanding of OR gene regulation and highlight important standing questions regarding the emerging specificity of ultra-long-range genomic interaction and the contribution of transcription and noncoding RNAs.
Collapse
Affiliation(s)
- Longzhi Tan
- Department of Neurobiology, Stanford University, Stanford, CA, USA. https://twitter.com/@tanlongzhi
| | - X Sunney Xie
- Biomedical Pioneering Innovation Center (BIOPIC), and School of Life Sciences, Peking University, Beijing, China; Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA. https://twitter.com/@XieSunney
| | - Stavros Lomvardas
- Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Zuckerman Mind, Brain, and Behavior Institute, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
2
|
Kanata E, Duffié R, Schulz EG. Establishment and maintenance of random monoallelic expression. Development 2024; 151:dev201741. [PMID: 38813842 PMCID: PMC11166465 DOI: 10.1242/dev.201741] [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: 05/31/2024]
Abstract
This Review elucidates the regulatory principles of random monoallelic expression by focusing on two well-studied examples: the X-chromosome inactivation regulator Xist and the olfactory receptor gene family. Although the choice of a single X chromosome or olfactory receptor occurs in different developmental contexts, common gene regulatory principles guide monoallelic expression in both systems. In both cases, an event breaks the symmetry between genetically and epigenetically identical copies of the gene, leading to the expression of one single random allele, stabilized through negative feedback control. Although many regulatory steps that govern the establishment and maintenance of monoallelic expression have been identified, key pieces of the puzzle are still missing. We provide an overview of the current knowledge and models for the monoallelic expression of Xist and olfactory receptors. We discuss their similarities and differences, and highlight open questions and approaches that could guide the study of other monoallelically expressed genes.
Collapse
Affiliation(s)
- Eleni Kanata
- Systems Epigenetics, Otto Warburg Laboratories, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Rachel Duffié
- Department of Biochemistry and Molecular Biophysics, Mortimer B. Zuckerman Mind, Brain, and Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Edda G. Schulz
- Systems Epigenetics, Otto Warburg Laboratories, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| |
Collapse
|
3
|
Yusuf N, Monahan K. Epigenetic programming of stochastic olfactory receptor choice. Genesis 2024; 62:e23593. [PMID: 38562011 PMCID: PMC11003729 DOI: 10.1002/dvg.23593] [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: 03/01/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
The mammalian sense of smell relies upon a vast array of receptor proteins to detect odorant compounds present in the environment. The proper deployment of these receptor proteins in olfactory sensory neurons is orchestrated by a suite of epigenetic processes that remodel the olfactory genes in differentiating neuronal progenitors. The goal of this review is to elucidate the central role of gene regulatory processes acting in neuronal progenitors of olfactory sensory neurons that lead to a singular expression of an odorant receptor in mature olfactory sensory neurons. We begin by describing the principal features of odorant receptor gene expression in mature olfactory sensory neurons. Next, we delineate our current understanding of how these features emerge from multiple gene regulatory mechanisms acting in neuronal progenitors. Finally, we close by discussing the key gaps in our understanding of how these regulatory mechanisms work and how they interact with each other over the course of differentiation.
Collapse
Affiliation(s)
- Nusrath Yusuf
- Division of Life Sciences-Molecular Biology and Biochemistry Department, Rutgers University-New Brunswick, New Brunswick, New Jersey, USA
| | - Kevin Monahan
- Division of Life Sciences-Molecular Biology and Biochemistry Department, Rutgers University-New Brunswick, New Brunswick, New Jersey, USA
| |
Collapse
|
4
|
Hussainy M, Korsching SI, Tresch A. Pseudotime analysis reveals novel regulatory factors for multigenic onset and monogenic transition of odorant receptor expression. Sci Rep 2022; 12:16183. [PMID: 36171231 PMCID: PMC9519747 DOI: 10.1038/s41598-022-20106-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 09/08/2022] [Indexed: 12/02/2022] Open
Abstract
During their maturation from horizontal basal stem cells, olfactory sensory neurons (OSNs) are known to select exactly one out of hundreds of olfactory receptors (ORs) and express it on their surface, a process called monogenic selection. Monogenic expression is preceded by a multigenic phase during which several OR genes are expressed in a single OSN. Here, we perform pseudotime analysis of a single cell RNA-Seq dataset of murine olfactory epithelium to precisely align the multigenic and monogenic expression phases with the cell types occurring during OSN differentiation. In combination with motif analysis of OR gene cluster-associated enhancer regions, we identify known and novel transcription (co-)factors (Ebf1, Lhx2, Ldb1, Fos and Ssbp2) and chromatin remodelers (Kdm1a, Eed and Zmynd8) associated with OR expression. The inferred temporal order of their activity suggests novel mechanisms contributing to multigenic OR expression and monogenic selection.
Collapse
Affiliation(s)
- Mohammad Hussainy
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University of Cologne, Cologne, Germany.,Institute of Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Sigrun I Korsching
- Institute of Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Achim Tresch
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University of Cologne, Cologne, Germany. .,Cologne Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany. .,Center for Data and Simulation Science, University of Cologne, Cologne, Germany.
| |
Collapse
|
5
|
Wang Y, Madhusudan S, Cotellessa L, Kvist J, Eskici N, Yellapragada V, Pulli K, Lund C, Vaaralahti K, Tuuri T, Giacobini P, Raivio T. Deciphering the Transcriptional Landscape of Human Pluripotent Stem Cell-Derived GnRH Neurons: The Role of Wnt Signaling in Patterning the Neural Fate. Stem Cells 2022; 40:1107-1121. [PMID: 36153707 PMCID: PMC9806769 DOI: 10.1093/stmcls/sxac069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/14/2022] [Indexed: 01/05/2023]
Abstract
Hypothalamic gonadotropin-releasing hormone (GnRH) neurons lay the foundation for human development and reproduction; however, the critical cell populations and the entangled mechanisms underlying the development of human GnRH neurons remain poorly understood. Here, by using our established human pluripotent stem cell-derived GnRH neuron model, we decoded the cellular heterogeneity and differentiation trajectories at the single-cell level. We found that a glutamatergic neuron population, which generated together with GnRH neurons, showed similar transcriptomic properties with olfactory sensory neuron and provided the migratory path for GnRH neurons. Through trajectory analysis, we identified a specific gene module activated along the GnRH neuron differentiation lineage, and we examined one of the transcription factors, DLX5, expression in human fetal GnRH neurons. Furthermore, we found that Wnt inhibition could increase DLX5 expression and improve the GnRH neuron differentiation efficiency through promoting neurogenesis and switching the differentiation fates of neural progenitors into glutamatergic neurons/GnRH neurons. Our research comprehensively reveals the dynamic cell population transition and gene regulatory network during GnRH neuron differentiation.
Collapse
Affiliation(s)
- Yafei Wang
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Shrinidhi Madhusudan
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ludovica Cotellessa
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Jouni Kvist
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Nazli Eskici
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Venkatram Yellapragada
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kristiina Pulli
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Carina Lund
- Folkhälsan Research Center, Helsinki, Finland
| | - Kirsi Vaaralahti
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland,New Children’s Hospital, Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
| | | | - Taneli Raivio
- Corresponding author: Taneli Raivio, Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
6
|
Abbas G, Tang S, Noble J, Lane RP. Olfactory receptor coding sequences cause silencing of episomal constructs in multiple cell lines. Mol Cell Neurosci 2021; 117:103681. [PMID: 34742908 PMCID: PMC8669572 DOI: 10.1016/j.mcn.2021.103681] [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: 12/29/2020] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 10/19/2022] Open
Abstract
The mammalian olfactory system consists of sensory neurons with specialized odorant-binding capability accomplished by mutually exclusive odorant receptor (OR) expression. Mutually exclusive OR expression is a complex multi-step process regulated by a number of cis and trans factors, including pan-silencing of all OR genes preceding the robust and stable expression of the one OR selected in each sensory neuron. We transfected two olfactory-placode-derived cell lines modeling immature odorant sensory neurons, as well as the GD25 fibroblast cell line, with episomes containing CMV-driven GFP and TK-driven hygromycin reporter genes. We inserted various coding sequences, along with an IRES, immediately upstream of the GFP gene to produce bicistronic mRNAs driven from the local CMV promoter. We found that the presence of several OR coding sequences resulted in significantly diminished episomal expression of GFP in all three cell lines. These findings suggest that OR coding sequences have intrinsic self-silencing capability that might facilitate mutually exclusive OR expression in olfactory sensory neurons by making it less likely that multiple ORs acquire an above-threshold level of expression at once.
Collapse
Affiliation(s)
- Ghazia Abbas
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT 06457, USA
| | - Spencer Tang
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT 06457, USA
| | - Joyce Noble
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT 06457, USA
| | - Robert P Lane
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT 06457, USA.
| |
Collapse
|
7
|
Nakano H, Kawai S, Ooki Y, Chiba T, Ishii C, Nozawa T, Utsuki H, Umemura M, Takahashi S, Takahashi Y. Functional validation of epitope-tagged ATF5 knock-in mice generated by improved genome editing of oviductal nucleic acid delivery (i-GONAD). Cell Tissue Res 2021; 385:239-249. [PMID: 33825962 DOI: 10.1007/s00441-021-03450-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/10/2021] [Indexed: 11/25/2022]
Abstract
Activating transcription factor 5 (ATF5) is a stress-responsive transcription factor that belongs to the cAMP response element-binding protein (CREB)/ATF family, and is essential for the differentiation and survival of sensory neurons in murine olfactory organs. However, the study of associated proteins and target genes for ATF5 has been hampered due to the limited availability of immunoprecipitation-grade ATF5 antibodies. To overcome this issue, we generated hemagglutinin (HA)-tag knock-in mice for ATF5 using CRISPR/Cas9-mediated genome editing with one-step electroporation in oviducts (i-GONAD). ATF5-HA fusion proteins were detected in the nuclei of immature and some mature olfactory and vomeronasal sensory neurons in the main olfactory epithelium and vomeronasal organ, respectively, as endogenous ATF5 proteins were expressed, and some ATF5-HA proteins were found to be phosphorylated. Chromatin immunoprecipitation (ChIP) experiments revealed that ATF5-HA bound to the CCAAT/enhancer-binding protein (C/EBP)-ATF response element site in the promotor region of receptor transporting protein 1 (Rtp1), a chaperone gene responsible for proper olfactory receptor expression. These knock-in mice may be used to examine the expression, localization, and protein-protein/-DNA interactions of endogenous ATF5 and, ultimately, the function of ATF5 in vivo.
Collapse
Affiliation(s)
- Haruo Nakano
- Laboratory of Environmental Molecular Physiology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1, Horinouchi, Hachioji, Tokyo, 192-0392, Japan.
| | - Shiori Kawai
- Laboratory of Environmental Molecular Physiology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1, Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Yusaku Ooki
- Laboratory of Environmental Molecular Physiology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1, Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Tomoki Chiba
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Chiharu Ishii
- Laboratory of Environmental Molecular Physiology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1, Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Takumi Nozawa
- Laboratory of Environmental Molecular Physiology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1, Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Hisako Utsuki
- Laboratory of Environmental Molecular Physiology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1, Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Mariko Umemura
- Laboratory of Environmental Molecular Physiology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1, Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Shigeru Takahashi
- Laboratory of Environmental Molecular Physiology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1, Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Yuji Takahashi
- Laboratory of Environmental Molecular Physiology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1, Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| |
Collapse
|
8
|
Leme Silva AG, Nagai MH, Nakahara TS, Malnic B. Genetic Background Effects on the Expression of an Odorant Receptor Gene. Front Cell Neurosci 2021; 15:646413. [PMID: 33716678 PMCID: PMC7947310 DOI: 10.3389/fncel.2021.646413] [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: 12/26/2020] [Accepted: 02/08/2021] [Indexed: 11/19/2022] Open
Abstract
There are more than 1000 odorant receptor (OR) genes in the mouse genome. Each olfactory sensory neuron expresses only one of these genes, in a monoallelic fashion. The transcript abundance of homologous OR genes vary between distinct mouse strains. Here we analyzed the expression of the OR gene Olfr17 (also named P2) in different genomic contexts. Olfr17 is expressed at higher levels in the olfactory epithelium from 129 mice than from C57BL/6 (B6) mice. However, we found that in P2-IRES-tauGFP knock-in mice, the transcript levels of the 129 Olfr17 allele are highly reduced when compared to the B6 Olfr17 allele. To address the mechanisms involved in this variation we compared the 5′ region sequence and DNA methylation patterns of the B6 and 129 Olfr17 alleles. Our results show that genetic variations in cis regulatory regions can lead to differential DNA methylation frequencies in these OR gene alleles. They also show that expression of the Olfr17 alleles is largely affected by the genetic background, and suggest that in knock-in mice, expression can be affected by epigenetic modifications in the region of the targeted locus.
Collapse
Affiliation(s)
| | | | | | - Bettina Malnic
- Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
9
|
Iwata T, Tomeoka S, Hirota J. A class I odorant receptor enhancer shares a functional motif with class II enhancers. Sci Rep 2021; 11:510. [PMID: 33436797 PMCID: PMC7804114 DOI: 10.1038/s41598-020-79980-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/15/2020] [Indexed: 11/09/2022] Open
Abstract
In the mouse, 129 functional class I odorant receptor (OR) genes reside in a ~ 3 megabase huge gene cluster on chromosome 7. The J element, a long-range cis-regulatory element governs the singular expression of class I OR genes by exerting its effect over the whole cluster. To elucidate the molecular mechanisms underlying class I-specific enhancer activity of the J element, we analyzed the J element sequence to determine the functional region and essential motif. The 430-bp core J element, that is highly conserved in mammalian species from the platypus to humans, contains a class I-specific conserved motif of AAACTTTTC, multiple homeodomain sites, and a neighboring O/E-like site, as in class II OR-enhancers. A series of transgenic reporter assays demonstrated that the class I-specific motif is not essential, but the 330-bp core J-H/O containing the homeodomain and O/E-like sites is necessary and sufficient for class I-specific enhancer activity. Further motif analysis revealed that one of homeodomain sequence is the Greek Islands composite motif of the adjacent homeodomain and O/E-like sequences, and mutations in the composite motif abolished or severely reduced class I-enhancer activity. Our results demonstrate that class I and class II enhancers share a functional motif for their enhancer activity.
Collapse
Affiliation(s)
- Tetsuo Iwata
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8501, Japan.,Biomaterial Analysis Division, Technical Department, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Satoshi Tomeoka
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Junji Hirota
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8501, Japan. .,Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan.
| |
Collapse
|
10
|
Wragg JW, Roos L, Vucenovic D, Cvetesic N, Lenhard B, Müller F. Embryonic tissue differentiation is characterized by transitions in cell cycle dynamic-associated core promoter regulation. Nucleic Acids Res 2020; 48:8374-8392. [PMID: 32619237 PMCID: PMC7470974 DOI: 10.1093/nar/gkaa563] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/10/2020] [Accepted: 06/19/2020] [Indexed: 12/30/2022] Open
Abstract
The core-promoter, a stretch of DNA surrounding the transcription start site (TSS), is a major integration-point for regulatory-signals controlling gene-transcription. Cellular differentiation is marked by divergence in transcriptional repertoire and cell-cycling behaviour between cells of different fates. The role promoter-associated gene-regulatory-networks play in development-associated transitions in cell-cycle-dynamics is poorly understood. This study demonstrates in a vertebrate embryo, how core-promoter variations define transcriptional output in cells transitioning from a proliferative to cell-lineage specifying phenotype. Assessment of cell proliferation across zebrafish embryo segmentation, using the FUCCI transgenic cell-cycle-phase marker, revealed a spatial and lineage-specific separation in cell-cycling behaviour. To investigate the role differential promoter usage plays in this process, cap-analysis-of-gene-expression (CAGE) was performed on cells segregated by cycling dynamics. This analysis revealed a dramatic increase in tissue-specific gene expression, concurrent with slowed cycling behaviour. We revealed a distinct sharpening in TSS utilization in genes upregulated in slowly cycling, differentiating tissues, associated with enhanced utilization of the TATA-box, in addition to Sp1 binding-sites. In contrast, genes upregulated in rapidly cycling cells carry broad distribution of TSS utilization, coupled with enrichment for the CCAAT-box. These promoter features appear to correspond to cell-cycle-dynamic rather than tissue/cell-lineage origin. Moreover, we observed genes with cell-cycle-dynamic-associated transitioning in TSS distribution and differential utilization of alternative promoters. These results demonstrate the regulatory role of core-promoters in cell-cycle-dependent transcription regulation, during embryo-development.
Collapse
Affiliation(s)
| | | | - Dunja Vucenovic
- Institute of Clinical Sciences and MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Nevena Cvetesic
- Institute of Clinical Sciences and MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Boris Lenhard
- Correspondence may also be addressed to Boris Lenhard. Tel: +44 20 3313 8353;
| | - Ferenc Müller
- To whom correspondence should be addressed. Tel: +44 121 414 2895;
| |
Collapse
|
11
|
A Group of Olfactory Receptor Alleles that Encode Full Length Proteins are Down-Regulated as Olfactory Sensory Neurons Mature. Sci Rep 2020; 10:1781. [PMID: 32020011 PMCID: PMC7000733 DOI: 10.1038/s41598-020-58779-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 01/21/2020] [Indexed: 11/08/2022] Open
Abstract
The family of olfactory receptors (ORs) subserves the sense of smell and includes both functional alleles and pseudogenes, the latter identified by mutations resulting in frame shift or premature truncation. During neuronal differentiation, nonfunctional ORs are expressed initially but then are switched out, and/or the olfactory sensory neurons (OSNs) expressing them die. We carried out a transcriptomic analysis of FACS-isolated cells from ΔSox2-eGFP, Neurog1-eGFP BAC and ΔOMP-eGFP strains of uninjured and olfactory bulbectomized transgenic mice that correspond to distinct stages in the progression from globose basal cell stem cells to fully mature OSNs. We analyzed the expression pattern of 1094 unique receptors across this progression and found that the vast majority were characterized by a typical and expected pattern of expression; i.e., levels of OR mRNA peaking in mature OSNs. However, 43 ORs, including several known pseudogenes, were different, such that mRNA expression declined in the mature OSNs relative to earlier stages. Protein and promoter sequence analysis of the atypical group did not uncover any obvious differences between them and more typical ORs. Nonetheless, the pattern of expression suggests that atypical ORs may be non-functional despite the lack of any obvious abnormality in the sequence analyses.
Collapse
|
12
|
Bertuzzi M, Tang D, Calligaris R, Vlachouli C, Finaurini S, Sanges R, Goldwurm S, Catalan M, Antonutti L, Manganotti P, Pizzolato G, Pezzoli G, Persichetti F, Carninci P, Gustincich S. A human minisatellite hosts an alternative transcription start site for NPRL3 driving its expression in a repeat number-dependent manner. Hum Mutat 2020; 41:807-824. [PMID: 31898848 DOI: 10.1002/humu.23974] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 11/16/2019] [Accepted: 12/24/2019] [Indexed: 12/21/2022]
Abstract
Minisatellites, also called variable number of tandem repeats (VNTRs), are a class of repetitive elements that may affect gene expression at multiple levels and have been correlated to disease. Their identification and role as expression quantitative trait loci (eQTL) have been limited by their absence in comparative genomic hybridization and single nucleotide polymorphisms arrays. By taking advantage of cap analysis of gene expression (CAGE), we describe a new example of a minisatellite hosting a transcription start site (TSS) which expression is dependent on the repeat number. It is located in the third intron of the gene nitrogen permease regulator like protein 3 (NPRL3). NPRL3 is a component of the GAP activity toward rags 1 protein complex that inhibits mammalian target of rapamycin complex 1 (mTORC1) activity and it is found mutated in familial focal cortical dysplasia and familial focal epilepsy. CAGE tags represent an alternative TSS identifying TAGNPRL3 messenger RNAs (mRNAs). TAGNPRL3 is expressed in red blood cells both at mRNA and protein levels, it interacts with its protein partner NPRL2 and its overexpression inhibits cell proliferation. This study provides an example of a minisatellite that is both a TSS and an eQTL as well as identifies a new VNTR that may modify mTORC1 activity.
Collapse
Affiliation(s)
| | - Dave Tang
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Japan
| | - Raffaella Calligaris
- Area of Neuroscience, SISSA, Trieste, Italy.,Department of Medical Sciences, Neurology Unit, University of Trieste, Trieste, Italy
| | | | - Sara Finaurini
- Area of Neuroscience, SISSA, Trieste, Italy.,Department of Health Sciences, Università del Piemonte Orientale and IRCAD, Novara, Italy
| | - Remo Sanges
- Area of Neuroscience, SISSA, Trieste, Italy.,Central RNA Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | | | - Mauro Catalan
- Department of Medical Sciences, Neurology Unit, University of Trieste, Trieste, Italy
| | - Lucia Antonutti
- Department of Medical Sciences, Neurology Unit, University of Trieste, Trieste, Italy
| | - Paolo Manganotti
- Department of Medical Sciences, Neurology Unit, University of Trieste, Trieste, Italy
| | - Gilberto Pizzolato
- Department of Medical Sciences, Neurology Unit, University of Trieste, Trieste, Italy
| | - Gianni Pezzoli
- Parkinson Institute, ASST G. Pini-CTO, ex ICP, Milan, Italy
| | - Francesca Persichetti
- Department of Health Sciences, Università del Piemonte Orientale and IRCAD, Novara, Italy
| | - Piero Carninci
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Japan.,Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Stefano Gustincich
- Area of Neuroscience, SISSA, Trieste, Italy.,Central RNA Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
13
|
Farber JE, Lane RP. Bioinformatics Discovery of Putative Enhancers within Mouse Odorant Receptor Gene Clusters. Chem Senses 2019; 44:705-720. [PMID: 31529021 DOI: 10.1093/chemse/bjz043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Olfactory neuronal function depends on the expression and proper regulation of odorant receptor (OR) genes. Previous studies have identified 54 putative intergenic enhancers within or flanking 40 mouse OR clusters. At least 2 of these putative enhancers have been shown to regulate the expression of a small subset of proximal OR genes. In recognition of the large size of the mouse OR gene family (~1400 OR genes distributed across multiple chromosomal loci), it is likely that there remain many additional not-as-yet discovered OR enhancers. We utilized 23 of the previously identified enhancers as a training set (TS) and designed an algorithm that combines a broad range of epigenetic criteria (histone-3-lysine-4 monomethylation, histone-3-lysine-79 trimethylation, histone-3-lysine-27 acetylation, and DNase hypersensitivity) and genetic criteria (cross-species sequence conservation and transcription-factor binding site enrichment) to more broadly search OR gene clusters for additional candidates. We identified 181 new candidate enhancers located at 58 (of 68) mouse OR loci, including 25 new candidates identified by stringent search criteria whose signal strengths are not significantly different from the 23 previously characterized OR enhancers used as the TS. Additionally, we compared OR enhancer versus generic enhancer features in order to evaluate likelihoods that new enhancer candidates specifically function in OR regulation. We found that features distinguishing OR-specific function are significantly more evident for enhancer candidates located within OR clusters as compared with those in flanking regions.
Collapse
Affiliation(s)
- James E Farber
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Robert P Lane
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| |
Collapse
|
14
|
Enomoto T, Nishida H, Iwata T, Fujita A, Nakayama K, Kashiwagi T, Hatanaka Y, Kondo H, Kajitani R, Itoh T, Ohmoto M, Matsumoto I, Hirota J. Bcl11b controls odorant receptor class choice in mice. Commun Biol 2019; 2:296. [PMID: 31396576 PMCID: PMC6685970 DOI: 10.1038/s42003-019-0536-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 07/09/2019] [Indexed: 11/24/2022] Open
Abstract
Each olfactory sensory neuron (OSN) expresses a single odorant receptor (OR) gene from the class I or class II repertoire in mice. The mechanisms that regulate OR class choice in OSNs remain unknown. Here, we show that the transcription factor Bcl11b determines the OR class to be expressed in OSNs. Both loss- and gain-of-function analyses demonstrate that class I is a default fate of OSNs and that Bcl11b dictates a class II OR choice by suppressing the effect of the J-element, a class I-OR enhancer. We further demonstrate that OSN-specific genetic manipulations of Bcl11b bias the OR class choice, generating mice with "class I-dominant" and "class II-dominant" noses, which display contrasting innate olfactory behaviors to two distinct aversive odorants. Overall, these findings reveal a unique transcriptional mechanism mediating a binary switch for OR class choice that is crucial to both the anatomical and functional organization of the olfactory system.
Collapse
Affiliation(s)
- Takayuki Enomoto
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Hidefumi Nishida
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Tetsuo Iwata
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Akito Fujita
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Kanako Nakayama
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Takahiro Kashiwagi
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Yasue Hatanaka
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Hiro Kondo
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Rei Kajitani
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Takehiko Itoh
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Makoto Ohmoto
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
- Monell Chemical Senses Center, Philadelphia, PA 19104 USA
| | | | - Junji Hirota
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| |
Collapse
|
15
|
Doulazmi M, Cros C, Dusart I, Trembleau A, Dubacq C. Alternative polyadenylation produces multiple 3' untranslated regions of odorant receptor mRNAs in mouse olfactory sensory neurons. BMC Genomics 2019; 20:577. [PMID: 31299892 PMCID: PMC6624953 DOI: 10.1186/s12864-019-5927-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/23/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Odorant receptor genes constitute the largest gene family in mammalian genomes and this family has been extensively studied in several species, but to date far less attention has been paid to the characterization of their mRNA 3' untranslated regions (3'UTRs). Given the increasing importance of UTRs in the understanding of RNA metabolism, and the growing interest in alternative polyadenylation especially in the nervous system, we aimed at identifying the alternative isoforms of odorant receptor mRNAs generated through 3'UTR variation. RESULTS We implemented a dedicated pipeline using IsoSCM instead of Cufflinks to analyze RNA-Seq data from whole olfactory mucosa of adult mice and obtained an extensive description of the 3'UTR isoforms of odorant receptor mRNAs. To validate our bioinformatics approach, we exhaustively analyzed the 3'UTR isoforms produced from 2 pilot genes, using molecular approaches including northern blot and RNA ligation mediated polyadenylation test. Comparison between datasets further validated the pipeline and confirmed the alternative polyadenylation patterns of odorant receptors. Qualitative and quantitative analyses of the annotated 3' regions demonstrate that 1) Odorant receptor 3'UTRs are longer than previously described in the literature; 2) More than 77% of odorant receptor mRNAs are subject to alternative polyadenylation, hence generating at least 2 detectable 3'UTR isoforms; 3) Splicing events in 3'UTRs are restricted to a limited subset of odorant receptor genes; and 4) Comparison between male and female data shows no sex-specific differences in odorant receptor 3'UTR isoforms. CONCLUSIONS We demonstrated for the first time that odorant receptor genes are extensively subject to alternative polyadenylation. This ground-breaking change to the landscape of 3'UTR isoforms of Olfr mRNAs opens new avenues for investigating their respective functions, especially during the differentiation of olfactory sensory neurons.
Collapse
Affiliation(s)
- Mohamed Doulazmi
- CNRS, Institut de Biologie Paris Seine, Biological adaptation and ageing, B2A, Sorbonne Université, F-75005 Paris, France
| | - Cyril Cros
- CNRS, INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, NPS, Sorbonne Université, F-75005 Paris, France
- Present Address: Columbia University, New York, NY 10027 USA
| | - Isabelle Dusart
- CNRS, INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, NPS, Sorbonne Université, F-75005 Paris, France
| | - Alain Trembleau
- CNRS, INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, NPS, Sorbonne Université, F-75005 Paris, France
| | - Caroline Dubacq
- CNRS, INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, NPS, Sorbonne Université, F-75005 Paris, France
| |
Collapse
|
16
|
Mohrhardt J, Nagel M, Fleck D, Ben-Shaul Y, Spehr M. Signal Detection and Coding in the Accessory Olfactory System. Chem Senses 2019; 43:667-695. [PMID: 30256909 PMCID: PMC6211456 DOI: 10.1093/chemse/bjy061] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In many mammalian species, the accessory olfactory system plays a central role in guiding behavioral and physiological responses to social and reproductive interactions. Because of its relatively compact structure and its direct access to amygdalar and hypothalamic nuclei, the accessory olfactory pathway provides an ideal system to study sensory control of complex mammalian behavior. During the last several years, many studies employing molecular, behavioral, and physiological approaches have significantly expanded and enhanced our understanding of this system. The purpose of the current review is to integrate older and newer studies to present an updated and comprehensive picture of vomeronasal signaling and coding with an emphasis on early accessory olfactory system processing stages. These include vomeronasal sensory neurons in the vomeronasal organ, and the circuitry of the accessory olfactory bulb. Because the overwhelming majority of studies on accessory olfactory system function employ rodents, this review is largely focused on this phylogenetic order, and on mice in particular. Taken together, the emerging view from both older literature and more recent studies is that the molecular, cellular, and circuit properties of chemosensory signaling along the accessory olfactory pathway are in many ways unique. Yet, it has also become evident that, like the main olfactory system, the accessory olfactory system also has the capacity for adaptive learning, experience, and state-dependent plasticity. In addition to describing what is currently known about accessory olfactory system function and physiology, we highlight what we believe are important gaps in our knowledge, which thus define exciting directions for future investigation.
Collapse
Affiliation(s)
- Julia Mohrhardt
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Aachen, Germany
| | - Maximilian Nagel
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Aachen, Germany
| | - David Fleck
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Aachen, Germany
| | - Yoram Ben-Shaul
- Department of Medical Neurobiology, School of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Marc Spehr
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
17
|
Tan L, Xie XS. A Near-Complete Spatial Map of Olfactory Receptors in the Mouse Main Olfactory Epithelium. Chem Senses 2018; 43:427-432. [PMID: 29796642 PMCID: PMC6454507 DOI: 10.1093/chemse/bjy030] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Different regions of the mammalian nose smell different odors. In the mouse olfactory system, spatially regulated expression of >1000 olfactory receptors (ORs) along the dorsomedial-ventrolateral (DV) axis forms a topological map in the main olfactory epithelium (MOE). However, the locations of most ORs along the DV axis are currently unknown. By sequencing mRNA of 12 isolated MOE pieces, we mapped out the DV locations-as quantified by "zone indices" on a scale of 1-5-of 1033 OR genes with an estimated error of 0.3 zone indices. Our map covered 81% of all intact OR genes and 99.4% of the total OR mRNA abundance. Spatial regulation tended to vary gradually along chromosomes. We further identified putative non-OR genes that may exhibit spatial expression along the DV axis.
Collapse
Affiliation(s)
- Longzhi Tan
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Systems Biology PhD Program, Harvard Medical School, Boston, MA, USA
| | - Xiaoliang Sunney Xie
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| |
Collapse
|
18
|
Monahan K, Schieren I, Cheung J, Mumbey-Wafula A, Monuki ES, Lomvardas S. Cooperative interactions enable singular olfactory receptor expression in mouse olfactory neurons. eLife 2017; 6. [PMID: 28933695 PMCID: PMC5608512 DOI: 10.7554/elife.28620] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 08/28/2017] [Indexed: 12/16/2022] Open
Abstract
The monogenic and monoallelic expression of only one out of >1000 mouse olfactory receptor (ORs) genes requires the formation of large heterochromatic chromatin domains that sequester the OR gene clusters. Within these domains, intergenic transcriptional enhancers evade heterochromatic silencing and converge into interchromosomal hubs that assemble over the transcriptionally active OR. The significance of this nuclear organization in OR choice remains elusive. Here, we show that transcription factors Lhx2 and Ebf specify OR enhancers by binding in a functionally cooperative fashion to stereotypically spaced motifs that defy heterochromatin. Specific displacement of Lhx2 and Ebf from OR enhancers resulted in pervasive, long-range, and trans downregulation of OR transcription, whereas pre-assembly of a multi-enhancer hub increased the frequency of OR choice in cis. Our data provide genetic support for the requirement and sufficiency of interchromosomal interactions in singular OR choice and generate general regulatory principles for stochastic, mutually exclusive gene expression programs.
Collapse
Affiliation(s)
- Kevin Monahan
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, United States
| | - Ira Schieren
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, United States
| | - Jonah Cheung
- New York Structural Biology Center, New York, United States
| | - Alice Mumbey-Wafula
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, United States
| | - Edwin S Monuki
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, United States
| | - Stavros Lomvardas
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, United States.,Department of Neuroscience, Columbia University, New York, United States.,Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, United States
| |
Collapse
|
19
|
A Review on Recent Computational Methods for Predicting Noncoding RNAs. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9139504. [PMID: 28553651 PMCID: PMC5434267 DOI: 10.1155/2017/9139504] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/06/2017] [Accepted: 02/15/2017] [Indexed: 12/20/2022]
Abstract
Noncoding RNAs (ncRNAs) play important roles in various cellular activities and diseases. In this paper, we presented a comprehensive review on computational methods for ncRNA prediction, which are generally grouped into four categories: (1) homology-based methods, that is, comparative methods involving evolutionarily conserved RNA sequences and structures, (2) de novo methods using RNA sequence and structure features, (3) transcriptional sequencing and assembling based methods, that is, methods designed for single and pair-ended reads generated from next-generation RNA sequencing, and (4) RNA family specific methods, for example, methods specific for microRNAs and long noncoding RNAs. In the end, we summarized the advantages and limitations of these methods and pointed out a few possible future directions for ncRNA prediction. In conclusion, many computational methods have been demonstrated to be effective in predicting ncRNAs for further experimental validation. They are critical in reducing the huge number of potential ncRNAs and pointing the community to high confidence candidates. In the future, high efficient mapping technology and more intrinsic sequence features (e.g., motif and k-mer frequencies) and structure features (e.g., minimum free energy, conserved stem-loop, or graph structures) are suggested to be combined with the next- and third-generation sequencing platforms to improve ncRNA prediction.
Collapse
|
20
|
Degl'Innocenti A, D'Errico A. Regulatory Features for Odorant Receptor Genes in the Mouse Genome. Front Genet 2017; 8:19. [PMID: 28270833 PMCID: PMC5318403 DOI: 10.3389/fgene.2017.00019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/08/2017] [Indexed: 12/31/2022] Open
Abstract
The odorant receptor genes, seven transmembrane receptor genes constituting the vastest mammalian gene multifamily, are expressed monogenically and monoallelicaly in each sensory neuron in the olfactory epithelium. This characteristic, often referred to as the one neuron-one receptor rule, is driven by mostly uncharacterized molecular dynamics, generally named odorant receptor gene choice. Much attention has been paid by the scientific community to the identification of sequences regulating the expression of odorant receptor genes within their loci, where related genes are usually arranged in genomic clusters. A number of studies identified transcription factor binding sites on odorant receptor promoter sequences. Similar binding sites were also found on a number of enhancers that regulate in cis their transcription, but have been proposed to form interchromosomal networks. Odorant receptor gene choice seems to occur via the local removal of strongly repressive epigenetic markings, put in place during the maturation of the sensory neuron on each odorant receptor locus. Here we review the fast-changing state of art for the study of regulatory features for odorant receptor genes.
Collapse
Affiliation(s)
- Andrea Degl'Innocenti
- Max Planck Institute of BiophysicsFrankfurt am Main, Germany; Cell and Developmental Biology Unit, Department of Biology, University of PisaPisa, Italy; Center for Micro-BioRobotics, Italian Institute of Technology, Sant'Anna School of Advanced StudiesPisa, Italy
| | - Anna D'Errico
- Max Planck Institute of Biophysics Frankfurt am Main, Germany
| |
Collapse
|
21
|
Lhx2 Determines Odorant Receptor Expression Frequency in Mature Olfactory Sensory Neurons. eNeuro 2016; 3:eN-NWR-0230-16. [PMID: 27822500 PMCID: PMC5086798 DOI: 10.1523/eneuro.0230-16.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 10/04/2016] [Accepted: 10/10/2016] [Indexed: 02/08/2023] Open
Abstract
A developmental program of epigenetic repression prepares each mammalian olfactory sensory neuron (OSN) to strongly express one allele from just one of hundreds of odorant receptor (OR) genes, but what completes this process of OR gene choice by driving the expression of this allele is incompletely understood. Conditional deletion experiments in mice demonstrate that Lhx2 is necessary for normal expression frequencies of nearly all ORs and all trace amine-associated receptors, irrespective of whether the deletion of Lhx2 is initiated in immature or mature OSNs. Given previous evidence that Lhx2 binds OR gene control elements, these findings indicate that Lhx2 is directly involved in driving OR expression. The data also support the conclusion that OR expression is necessary to allow immature OSNs to complete differentiation and become mature. In contrast to the robust effects of conditional deletion of Lhx2, the loss of Emx2 has much smaller effects and more often causes increased expression frequencies. Lhx2:Emx2 double mutants show opposing effects on Olfr15 expression that reveal independent effects of these two transcription factors. While Lhx2 is necessary for OR expression that supports OR gene choice, Emx2 can act differently; perhaps by helping to control the availability of OR genes for expression.
Collapse
|
22
|
Marcuzzi F, Zucchelli S, Bertuzzi M, Santoro C, Tell G, Carninci P, Gustincich S. Isoforms of the Erythropoietin receptor in dopaminergic neurons of the Substantia Nigra. J Neurochem 2016; 139:596-609. [PMID: 27488413 DOI: 10.1111/jnc.13757] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 07/15/2016] [Accepted: 07/18/2016] [Indexed: 11/29/2022]
Abstract
Erythropoietin receptor (EpoR) regulates erythrocytes differentiation in blood. In the brain, EpoR has been shown to protect several neuronal cell types from cell death, including the A9 dopaminergic neurons (DA) of the Substantia Nigra (SN). These cells form the nigrostriatal pathway and are devoted to the control of postural reflexes and voluntary movements. Selective degeneration of A9 DA neurons leads to Parkinson's disease. By the use of nanoCAGE, a technology that allows the identification of Transcription Start Sites (TSSs) at a genome-wide level, we have described the promoter-level expression atlas of mouse A9 DA neurons purified with Laser Capture Microdissection (LCM). Here, we identify mRNA variants of the Erythropoietin Receptor (DA-EpoR) transcribed from alternative TSSs. Experimental validation and full-length cDNA cloning is integrated with gene expression analysis in the FANTOM5 database. In DA neurons, the EpoR gene encodes for a N-terminal truncated receptor. Based on STAT5 phosphorylation assays, we show that the new variant of N-terminally truncated EpoR acts as decoy when co-expressed with the full-length form. A similar isoform is also found in human. This work highlights new complexities in the regulation of Erythropoietin (EPO) signaling in the brain.
Collapse
Affiliation(s)
| | - Silvia Zucchelli
- Area of Neuroscience, SISSA, Trieste, Italy.,Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | | | - Claudio Santoro
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Gianluca Tell
- Department of Medical and Biological Sciences (DSMB), University of Udine, Udine, Italy
| | - Piero Carninci
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Japan
| | | |
Collapse
|
23
|
Olender T, Keydar I, Pinto JM, Tatarskyy P, Alkelai A, Chien MS, Fishilevich S, Restrepo D, Matsunami H, Gilad Y, Lancet D. The human olfactory transcriptome. BMC Genomics 2016; 17:619. [PMID: 27515280 PMCID: PMC4982115 DOI: 10.1186/s12864-016-2960-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 07/21/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Olfaction is a versatile sensory mechanism for detecting thousands of volatile odorants. Although molecular basis of odorant signaling is relatively well understood considerable gaps remain in the complete charting of all relevant gene products. To address this challenge, we applied RNAseq to four well-characterized human olfactory epithelial samples and compared the results to novel and published mouse olfactory epithelium as well as 16 human control tissues. RESULTS We identified 194 non-olfactory receptor (OR) genes that are overexpressed in human olfactory tissues vs. CONTROLS The highest overexpression is seen for lipocalins and bactericidal/permeability-increasing (BPI)-fold proteins, which in other species include secreted odorant carriers. Mouse-human discordance in orthologous lipocalin expression suggests different mammalian evolutionary paths in this family. Of the overexpressed genes 36 have documented olfactory function while for 158 there is little or no previous such functional evidence. The latter group includes GPCRs, neuropeptides, solute carriers, transcription factors and biotransformation enzymes. Many of them may be indirectly implicated in sensory function, and ~70 % are over expressed also in mouse olfactory epithelium, corroborating their olfactory role. Nearly 90 % of the intact OR repertoire, and ~60 % of the OR pseudogenes are expressed in the olfactory epithelium, with the latter showing a 3-fold lower expression. ORs transcription levels show a 1000-fold inter-paralog variation, as well as significant inter-individual differences. We assembled 160 transcripts representing 100 intact OR genes. These include 1-4 short 5' non-coding exons with considerable alternative splicing and long last exons that contain the coding region and 3' untranslated region of highly variable length. Notably, we identified 10 ORs with an intact open reading frame but with seemingly non-functional transcripts, suggesting a yet unreported OR pseudogenization mechanism. Analysis of the OR upstream regions indicated an enrichment of the homeobox family transcription factor binding sites and a consensus localization of a specific transcription factor binding site subfamily (Olf/EBF). CONCLUSIONS We provide an overview of expression levels of ORs and auxiliary genes in human olfactory epithelium. This forms a transcriptomic view of the entire OR repertoire, and reveals a large number of over-expressed uncharacterized human non-receptor genes, providing a platform for future discovery.
Collapse
Affiliation(s)
- Tsviya Olender
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| | - Ifat Keydar
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Jayant M Pinto
- Section of Otolaryngology-Head and Neck Surgery, University of Chicago, Chicago, IL, USA
| | - Pavlo Tatarskyy
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Anna Alkelai
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ming-Shan Chien
- Department of Molecular Genetics and Microbiology, Department of Neurobiology, Duke Institute for Brain Sciences, Duke University Medical Center, Durham, NC, USA
| | - Simon Fishilevich
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Diego Restrepo
- Department of Cell and Developmental Biology, Neuroscience Program, and Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Hiroaki Matsunami
- Department of Molecular Genetics and Microbiology, Department of Neurobiology, Duke Institute for Brain Sciences, Duke University Medical Center, Durham, NC, USA
| | - Yoav Gilad
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Doron Lancet
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
24
|
Wragg J, Müller F. Transcriptional Regulation During Zygotic Genome Activation in Zebrafish and Other Anamniote Embryos. ADVANCES IN GENETICS 2016; 95:161-94. [PMID: 27503357 DOI: 10.1016/bs.adgen.2016.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Embryo development commences with the fusion of two terminally differentiated haploid gametes into the totipotent fertilized egg, which through a series of major cellular and molecular transitions generate a pluripotent cell mass. The activation of the zygotic genome occurs during the so-called maternal to zygotic transition and prepares the embryo for zygotic takeover from maternal factors, in the control of the development of cellular lineages during differentiation. Recent advances in next generation sequencing technologies have allowed the dissection of the genomic and epigenomic processes mediating this transition. These processes include reorganization of the chromatin structure to a transcriptionally permissive state, changes in composition and function of structural and regulatory DNA-binding proteins, and changeover of the transcriptome as it is overhauled from that deposited by the mother in the oocyte to a zygotically transcribed complement. Zygotic genome activation in zebrafish occurs 10 cell cycles after fertilization and provides an ideal experimental platform for elucidating the temporal sequence and dynamics of establishment of a transcriptionally active chromatin state and helps in identifying the determinants of transcription activation at polymerase II transcribed gene promoters. The relatively large number of pluripotent cells generated by the fast cell divisions before zygotic transcription provides sufficient biomass for next generation sequencing technology approaches to establish the temporal dynamics of events and suggest causative relationship between them. However, genomic and genetic technologies need to be improved further to capture the earliest events in development, where cell number is a limiting factor. These technologies need to be complemented with precise, inducible genetic interference studies using the latest genome editing tools to reveal the function of candidate determinants and to confirm the predictions made by classic embryological tools and genome-wide assays. In this review we summarize recent advances in the characterization of epigenetic regulation, transcription control, and gene promoter function during zygotic genome activation and how they fit with old models for the mechanisms of the maternal to zygotic transition. This review will focus on the zebrafish embryo but draw comparisons with other vertebrate model systems and refer to invertebrate models where informative.
Collapse
Affiliation(s)
- J Wragg
- University of Birmingham, Birmingham, United Kingdom
| | - F Müller
- University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
25
|
Lomvardas S, Maniatis T. Histone and DNA Modifications as Regulators of Neuronal Development and Function. Cold Spring Harb Perspect Biol 2016; 8:8/7/a024208. [PMID: 27371659 DOI: 10.1101/cshperspect.a024208] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
DNA and histone modifications, together with constraints imposed by nuclear architecture, contribute to the transcriptional regulatory landscape of the nervous system. Here, we provide select examples showing how these regulatory layers, often referred to as epigenetic, contribute to neuronal differentiation and function. We describe the interplay between DNA methylation and Polycomb-mediated repression during neuronal differentiation, the role of DNA methylation and long-range enhancer-promoter interactions in Protocadherin promoter choice, and the contribution of heterochromatic silencing and nuclear organization in singular olfactory receptor expression. Finally, we explain how the activity-dependent expression of a histone variant determines the longevity of olfactory sensory neurons.
Collapse
Affiliation(s)
- Stavros Lomvardas
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York 10032
| | - Tom Maniatis
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York 10032
| |
Collapse
|
26
|
Chandler JD, Wongtrakool C, Banton SA, Li S, Orr ML, Barr DB, Neujahr DC, Sutliff RL, Go YM, Jones DP. Low-dose oral cadmium increases airway reactivity and lung neuronal gene expression in mice. Physiol Rep 2016; 4:e12821. [PMID: 27401458 PMCID: PMC4945833 DOI: 10.14814/phy2.12821] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 05/15/2016] [Indexed: 01/05/2023] Open
Abstract
Inhalation of cadmium (Cd) is associated with lung diseases, but less is known concerning pulmonary effects of Cd found in the diet. Cd has a decades-long half-life in humans and significant bioaccumulation occurs with chronic dietary intake. We exposed mice to low-dose CdCl2 (10 mg/L in drinking water) for 20 weeks, which increased lung Cd to a level similar to that of nonoccupationally exposed adult humans. Cd-treated mice had increased airway hyperresponsiveness to methacholine challenge, and gene expression array showed that Cd altered the abundance of 443 mRNA transcripts in mouse lung. In contrast to higher doses, low-dose Cd did not elicit increased metallothionein transcripts in lung. To identify pathways most affected by Cd, gene set enrichment of transcripts was analyzed. Results showed that major inducible targets of low-dose Cd were neuronal receptors represented by enriched olfactory, glutamatergic, cholinergic, and serotonergic gene sets. Olfactory receptors regulate chemosensory function and airway hypersensitivity, and these gene sets were the most enriched. Targeted metabolomics analysis showed that Cd treatment also increased metabolites in pathways of glutamatergic (glutamate), serotonergic (tryptophan), cholinergic (choline), and catecholaminergic (tyrosine) receptors in the lung tissue. Protein abundance measurements showed that the glutamate receptor GRIN2A was increased in mouse lung tissue. Together, these results show that in mice, oral low-dose Cd increased lung Cd to levels comparable to humans, increased airway hyperresponsiveness and disrupted neuronal pathways regulating bronchial tone. Therefore, dietary Cd may promote or worsen airway hyperresponsiveness in multiple lung diseases including asthma.
Collapse
Affiliation(s)
- Joshua D Chandler
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, Georgia
| | - Cherry Wongtrakool
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, Georgia Atlanta VA Medical Center, Decatur, Georgia
| | - Sophia A Banton
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, Georgia
| | - Shuzhao Li
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, Georgia
| | - Michael L Orr
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, Georgia
| | - Dana Boyd Barr
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - David C Neujahr
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, Georgia
| | - Roy L Sutliff
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, Georgia Atlanta VA Medical Center, Decatur, Georgia
| | - Young-Mi Go
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, Georgia
| | - Dean P Jones
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
27
|
Romano O, Peano C, Tagliazucchi GM, Petiti L, Poletti V, Cocchiarella F, Rizzi E, Severgnini M, Cavazza A, Rossi C, Pagliaro P, Ambrosi A, Ferrari G, Bicciato S, De Bellis G, Mavilio F, Miccio A. Transcriptional, epigenetic and retroviral signatures identify regulatory regions involved in hematopoietic lineage commitment. Sci Rep 2016; 6:24724. [PMID: 27095295 PMCID: PMC4837375 DOI: 10.1038/srep24724] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 04/04/2016] [Indexed: 12/21/2022] Open
Abstract
Genome-wide approaches allow investigating the molecular circuitry wiring the genetic and epigenetic programs of human somatic stem cells. Hematopoietic stem/progenitor cells (HSPC) give rise to the different blood cell types; however, the molecular basis of human hematopoietic lineage commitment is poorly characterized. Here, we define the transcriptional and epigenetic profile of human HSPC and early myeloid and erythroid progenitors by a combination of Cap Analysis of Gene Expression (CAGE), ChIP-seq and Moloney leukemia virus (MLV) integration site mapping. Most promoters and transcripts were shared by HSPC and committed progenitors, while enhancers and super-enhancers consistently changed upon differentiation, indicating that lineage commitment is essentially regulated by enhancer elements. A significant fraction of CAGE promoters differentially expressed upon commitment were novel, harbored a chromatin enhancer signature, and may identify promoters and transcribed enhancers driving cell commitment. MLV-targeted genomic regions co-mapped with cell-specific active enhancers and super-enhancers. Expression analyses, together with an enhancer functional assay, indicate that MLV integration can be used to identify bona fide developmentally regulated enhancers. Overall, this study provides an overview of transcriptional and epigenetic changes associated to HSPC lineage commitment, and a novel signature for regulatory elements involved in cell identity.
Collapse
Affiliation(s)
- Oriana Romano
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy.,INSERM UMR 1163, Laboratory of chromatin and gene regulation during development, Paris, France
| | - Clelia Peano
- Institute of Biomedical Technologies, CNR, Milan, Italy
| | | | - Luca Petiti
- Institute of Biomedical Technologies, CNR, Milan, Italy
| | | | | | - Ermanno Rizzi
- Institute of Biomedical Technologies, CNR, Milan, Italy.,Telethon Foundation, Milan, Italy
| | | | - Alessia Cavazza
- Dana Farber Cancer Institute, Harvard Medical School, Boston, US
| | - Claudia Rossi
- San Raffaele-Telethon Institute for Gene Therapy (TIGET), San Raffaele Scientific Institute, Milan, Italy
| | - Pasqualepaolo Pagliaro
- Az. Osp. Policlinico Universitario di Bologna, Policlinico S. Orsola-Malpighi, Unità Operativa di Immunoematologia e Trasfusionale, Bologna, Italy
| | | | - Giuliana Ferrari
- San Raffaele-Telethon Institute for Gene Therapy (TIGET), San Raffaele Scientific Institute, Milan, Italy.,Vita Salute San Raffaele University, Milan, Italy
| | - Silvio Bicciato
- Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Fulvio Mavilio
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Genethon, Evry, France
| | - Annarita Miccio
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.,INSERM UMR 1163, Laboratory of chromatin and gene regulation during development, Paris, France.,Paris Descartes, Sorbonne Paris Cité University, Imagine Institute, Paris, France
| |
Collapse
|
28
|
Degl'Innocenti A, Parrilla M, Harr B, Teschke M. The Mouse Solitary Odorant Receptor Gene Promoters as Models for the Study of Odorant Receptor Gene Choice. PLoS One 2016; 11:e0144698. [PMID: 26794459 PMCID: PMC4721658 DOI: 10.1371/journal.pone.0144698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/23/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In vertebrates, several anatomical regions located within the nasal cavity mediate olfaction. Among these, the main olfactory epithelium detects most conventional odorants. Olfactory sensory neurons, provided with cilia exposed to the air, detect volatile chemicals via an extremely large family of seven-transmembrane chemoreceptors named odorant receptors. Their genes are expressed in a monogenic and monoallelic fashion: a single allele of a single odorant receptor gene is transcribed in a given mature neuron, through a still uncharacterized molecular mechanism known as odorant receptor gene choice. AIM Odorant receptor genes are typically arranged in genomic clusters, but a few are isolated (we call them solitary) from the others within a region broader than 1 Mb upstream and downstream with respect to their transcript's coordinates. The study of clustered genes is problematic, because of redundancy and ambiguities in their regulatory elements: we propose to use the solitary genes as simplified models to understand odorant receptor gene choice. PROCEDURES Here we define number and identity of the solitary genes in the mouse genome (C57BL/6J), and assess the conservation of the solitary status in some mammalian orthologs. Furthermore, we locate their putative promoters, predict their homeodomain binding sites (commonly present in the promoters of odorant receptor genes) and compare candidate promoter sequences with those of wild-caught mice. We also provide expression data from histological sections. RESULTS In the mouse genome there are eight intact solitary genes: Olfr19 (M12), Olfr49, Olfr266, Olfr267, Olfr370, Olfr371, Olfr466, Olfr1402; five are conserved as solitary in rat. These genes are all expressed in the main olfactory epithelium of three-day-old mice. The C57BL/6J candidate promoter of Olfr370 has considerably varied compared to its wild-type counterpart. Within the putative promoter for Olfr266 a homeodomain binding site is predicted. As a whole, our findings favor Olfr266 as a model gene to investigate odorant receptor gene choice.
Collapse
Affiliation(s)
- Andrea Degl'Innocenti
- Max-Planck-Institut für Biophysik, Frankfurt am Main, Germany
- Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, Pisa, Italy
| | - Marta Parrilla
- Max-Planck-Institut für Biophysik, Frankfurt am Main, Germany
| | - Bettina Harr
- Abteilung Evolutionsgenetik, Max-Planck-Institut für Evolutionsbiologie, Plön, Germany
| | - Meike Teschke
- Abteilung Evolutionsgenetik, Max-Planck-Institut für Evolutionsbiologie, Plön, Germany
| |
Collapse
|
29
|
Kim JM, Ren D, Reverter A, Roura E. A regulatory gene network related to the porcine umami taste receptor (TAS1R1/TAS1R3). Anim Genet 2015; 47:114-9. [PMID: 26554867 DOI: 10.1111/age.12374] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2015] [Indexed: 11/28/2022]
Abstract
Taste perception plays an important role in the mediation of food choices in mammals. The first porcine taste receptor genes identified, sequenced and characterized, TAS1R1 and TAS1R3, were related to the dimeric receptor for umami taste. However, little is known about their regulatory network. The objective of this study was to unfold the genetic network involved in porcine umami taste perception. We performed a meta-analysis of 20 gene expression studies spanning 480 porcine microarray chips and screened 328 taste-related genes by selective mining steps among the available 12,320 genes. A porcine umami taste-specific regulatory network was constructed based on the normalized coexpression data of the 328 genes across 27 tissues. From the network, we revealed the 'taste module' and identified a coexpression cluster for the umami taste according to the first connector with the TAS1R1/TAS1R3 genes. Our findings identify several taste-related regulatory genes and extend previous genetic background of porcine umami taste.
Collapse
Affiliation(s)
- J M Kim
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation Hartley Teakle 83, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - D Ren
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation Hartley Teakle 83, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - A Reverter
- CSIRO Agriculture Flagship, Queensland Bioscience Precinct, 306 Carmody Road, St Lucia, Queensland, 4067, Australia
| | - E Roura
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation Hartley Teakle 83, The University of Queensland, St Lucia, Queensland, 4072, Australia
| |
Collapse
|
30
|
Abstract
The sense of smell collects vital information about the environment by detecting a multitude of chemical odorants. Breadth and sensitivity are provided by a huge number of chemosensory receptor proteins, including more than 1,400 olfactory receptors (ORs). Organizing the sensory information generated by these receptors so that it can be processed and evaluated by the central nervous system is a major challenge. This challenge is overcome by monogenic and monoallelic expression of OR genes. The single OR expressed by each olfactory sensory neuron determines the neuron's odor sensitivity and the axonal connections it will make to downstream neurons in the olfactory bulb. The expression of a single OR per neuron is accomplished by coupling a slow chromatin-mediated activation process to a fast negative-feedback signal that prevents activation of additional ORs. Singular OR activation is likely orchestrated by a network of interchromosomal enhancer interactions and large-scale changes in nuclear architecture.
Collapse
Affiliation(s)
- Kevin Monahan
- Department of Biochemistry and Molecular Biophysics, Department of Neuroscience, and Mortimer B. Zuckerman Mind, Brain, and Behavior Institute, Columbia University, New York, NY 10032; ,
| | - Stavros Lomvardas
- Department of Biochemistry and Molecular Biophysics, Department of Neuroscience, and Mortimer B. Zuckerman Mind, Brain, and Behavior Institute, Columbia University, New York, NY 10032; ,
| |
Collapse
|
31
|
Alqadah A, Hsieh YW, Vidal B, Chang C, Hobert O, Chuang CF. Postmitotic diversification of olfactory neuron types is mediated by differential activities of the HMG-box transcription factor SOX-2. EMBO J 2015; 34:2574-89. [PMID: 26341465 DOI: 10.15252/embj.201592188] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/06/2015] [Indexed: 12/23/2022] Open
Abstract
Diversification of neuron classes is essential for functions of the olfactory system, but the underlying mechanisms that generate individual olfactory neuron types are only beginning to be understood. Here we describe a role of the highly conserved HMG-box transcription factor SOX-2 in postmitotic specification and alternative differentiation of the Caenorhabditis elegans AWC and AWB olfactory neurons. We show that SOX-2 partners with different transcription factors to diversify postmitotic olfactory cell types. SOX-2 functions cooperatively with the OTX/OTD transcription factor CEH-36 to specify an AWC "ground state," and functions with the LIM homeodomain factor LIM-4 to suppress this ground state and drive an AWB identity instead. Our findings provide novel insights into combinatorial codes that drive terminal differentiation programs in the nervous system and reveal a biological function of the deeply conserved Sox2 protein that goes beyond its well-known role in stem cell biology.
Collapse
Affiliation(s)
- Amel Alqadah
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA Molecular and Developmental Biology Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Yi-Wen Hsieh
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Berta Vidal
- Department of Biological Sciences, Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute Columbia University, New York, NY, USA
| | - Chieh Chang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Oliver Hobert
- Department of Biological Sciences, Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute Columbia University, New York, NY, USA
| | - Chiou-Fen Chuang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
32
|
von der Weid B, Rossier D, Lindup M, Tuberosa J, Widmer A, Col JD, Kan C, Carleton A, Rodriguez I. Large-scale transcriptional profiling of chemosensory neurons identifies receptor-ligand pairs in vivo. Nat Neurosci 2015; 18:1455-63. [PMID: 26322926 DOI: 10.1038/nn.4100] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/04/2015] [Indexed: 12/11/2022]
Abstract
In mammals, olfactory perception is based on the combinatorial activation of G protein-coupled receptors. Identifying the full repertoire of receptors activated by a given odorant in vivo, a quest that has been hampered for over 20 years by technical difficulties, would represent an important step in deciphering the rules governing chemoperception. We found that odorants induced a fast and reversible concentration-dependent decrease in the transcription of genes corresponding to activated receptors in intact mice. On the basis of this finding, we developed a large-scale transcriptomic approach to uncover receptor-ligand pairs in vivo. We identified the mouse and rat odorant receptor signatures corresponding to specific odorants. Finally, we found that this approach, which can be used for species for which no genomic sequence is available, is also applicable to non-vertebrate species such as Drosophila.
Collapse
Affiliation(s)
- Benoît von der Weid
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | - Daniel Rossier
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | - Matti Lindup
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | - Joël Tuberosa
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | - Alexandre Widmer
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | - Julien Dal Col
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | - Chenda Kan
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | - Alan Carleton
- Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland.,Department of Basic Neurosciences, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Ivan Rodriguez
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| |
Collapse
|
33
|
Poletti V, Delli Carri A, Malagoli Tagliazucchi G, Faedo A, Petiti L, Mazza EMC, Peano C, De Bellis G, Bicciato S, Miccio A, Cattaneo E, Mavilio F. Genome-Wide Definition of Promoter and Enhancer Usage during Neural Induction of Human Embryonic Stem Cells. PLoS One 2015; 10:e0126590. [PMID: 25978676 PMCID: PMC4433211 DOI: 10.1371/journal.pone.0126590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/06/2015] [Indexed: 11/21/2022] Open
Abstract
Genome-wide mapping of transcriptional regulatory elements is an essential tool for understanding the molecular events orchestrating self-renewal, commitment and differentiation of stem cells. We combined high-throughput identification of transcription start sites with genome-wide profiling of histones modifications to map active promoters and enhancers in embryonic stem cells (ESCs) induced to neuroepithelial-like stem cells (NESCs). Our analysis showed that most promoters are active in both cell types while approximately half of the enhancers are cell-specific and account for most of the epigenetic changes occurring during neural induction, and most likely for the modulation of the promoters to generate cell-specific gene expression programs. Interestingly, the majority of the promoters activated or up-regulated during neural induction have a “bivalent” histone modification signature in ESCs, suggesting that developmentally-regulated promoters are already poised for transcription in ESCs, which are apparently pre-committed to neuroectodermal differentiation. Overall, our study provides a collection of differentially used enhancers, promoters, transcription starts sites, protein-coding and non-coding RNAs in human ESCs and ESC-derived NESCs, and a broad, genome-wide description of promoter and enhancer usage and of gene expression programs characterizing the transition from a pluripotent to a neural-restricted cell fate.
Collapse
Affiliation(s)
- Valentina Poletti
- Division of Genetics and Cell Biology, Scientific Institute H. San Raffaele, Milan, Italy
- Genethon, Evry, France
| | | | | | - Andrea Faedo
- Department of Biosciences, University of Milano, Milan, Italy
| | - Luca Petiti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Clelia Peano
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Gianluca De Bellis
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Annarita Miccio
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Imagine Institute, Paris, France
| | - Elena Cattaneo
- Department of Biosciences, University of Milano, Milan, Italy
| | - Fulvio Mavilio
- Genethon, Evry, France
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- * E-mail:
| |
Collapse
|
34
|
Abstract
The senses provide a means by which data on the physical and chemical properties of the environment may be collected and meaningfully interpreted. Sensation begins at the periphery, where a multitude of different sensory cell types are activated by environmental stimuli as different as photons and odorant molecules. Stimulus sensitivity is due to expression of different cell surface sensory receptors, and therefore the receptive field of each sense is defined by the aggregate of expressed receptors in each sensory tissue. Here, we review current understanding on patterns of expression and modes of regulation of sensory receptors.
Collapse
|
35
|
Jafari S, Alenius M. Cis-regulatory mechanisms for robust olfactory sensory neuron class-restricted odorant receptor gene expression in Drosophila. PLoS Genet 2015; 11:e1005051. [PMID: 25760344 PMCID: PMC4356613 DOI: 10.1371/journal.pgen.1005051] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/02/2015] [Indexed: 12/26/2022] Open
Abstract
Odor perception requires that each olfactory sensory neuron (OSN) class continuously express a single odorant receptor (OR) regardless of changes in the environment. However, little is known about the control of the robust, class-specific OR expression involved. Here, we investigate the cis-regulatory mechanisms and components that generate robust and OSN class-specific OR expression in Drosophila. Our results demonstrate that the spatial restriction of expression to a single OSN class is directed by clusters of transcription-factor DNA binding motifs. Our dissection of motif clusters of differing complexity demonstrates that structural components such as motif overlap and motif order integrate transcription factor combinations and chromatin status to form a spatially restricted pattern. We further demonstrate that changes in metabolism or temperature perturb the function of complex clusters. We show that the cooperative regulation between motifs around and within the cluster generates robust, class-specific OR expression. Our neurons can become over a hundred years old. Even if neurons are restructured and remodeled by their constant work of receiving, storing and sending information, they stay devoted to one single task and retain their identity for their whole life. How a neuron keeps its identity is not well understood. In the olfactory system, the identity of the olfactory sensory neuron (OSN) is a result of the expression of a single odorant receptor (OR) from a large receptor gene repertoire in the genome. Neurons that share an expressed receptor make a functional class. Here, we identify clusters of transcription factor binding motifs to be the smallest unit that drive expression in a single olfactory sensory neuron class. We further demonstrate that it is the structure of the cluster that determines the class specific expression. However, environmental stress, such as temperature changes or starvation, destabilizes the expression produced by the cluster. Our results demonstrate that stable expression is generated from redundant motifs outside the cluster and suggest that cooperative regulation generates robust expression of the genes that determine neuronal identity and function.
Collapse
Affiliation(s)
- Shadi Jafari
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Mattias Alenius
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- * E-mail:
| |
Collapse
|
36
|
Persuy MA, Sanz G, Tromelin A, Thomas-Danguin T, Gibrat JF, Pajot-Augy E. Mammalian olfactory receptors: molecular mechanisms of odorant detection, 3D-modeling, and structure-activity relationships. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 130:1-36. [PMID: 25623335 DOI: 10.1016/bs.pmbts.2014.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This chapter describes the main characteristics of olfactory receptor (OR) genes of vertebrates, including generation of this large multigenic family and pseudogenization. OR genes are compared in relation to evolution and among species. OR gene structure and selection of a given gene for expression in an olfactory sensory neuron (OSN) are tackled. The specificities of OR proteins, their expression, and their function are presented. The expression of OR proteins in locations other than the nasal cavity is regulated by different mechanisms, and ORs display various additional functions. A conventional olfactory signal transduction cascade is observed in OSNs, but individual ORs can also mediate different signaling pathways, through the involvement of other molecular partners and depending on the odorant ligand encountered. ORs are engaged in constitutive dimers. Ligand binding induces conformational changes in the ORs that regulate their level of activity depending on odorant dose. When present, odorant binding proteins induce an allosteric modulation of OR activity. Since no 3D structure of an OR has been yet resolved, modeling has to be performed using the closest G-protein-coupled receptor 3D structures available, to facilitate virtual ligand screening using the models. The study of odorant binding modes and affinities may infer best-bet OR ligands, to be subsequently checked experimentally. The relationship between spatial and steric features of odorants and their activity in terms of perceived odor quality are also fields of research that development of computing tools may enhance.
Collapse
Affiliation(s)
- Marie-Annick Persuy
- INRA UR 1197 NeuroBiologie de l'Olfaction, Domaine de Vilvert, Jouy-en-Josas, France
| | - Guenhaël Sanz
- INRA UR 1197 NeuroBiologie de l'Olfaction, Domaine de Vilvert, Jouy-en-Josas, France
| | - Anne Tromelin
- INRA UMR 1129 Flaveur, Vision et Comportement du Consommateur, Dijon, France
| | | | - Jean-François Gibrat
- INRA UR1077 Mathématique Informatique et Génome, Domaine de Vilvert, Jouy-en-Josas, France
| | - Edith Pajot-Augy
- INRA UR 1197 NeuroBiologie de l'Olfaction, Domaine de Vilvert, Jouy-en-Josas, France.
| |
Collapse
|
37
|
Abstract
The olfactory (OR) and vomeronasal receptor (VR) repertoires are collectively encoded by 1700 genes and pseudogenes in the mouse genome. Most OR and VR genes were identified by comparative genomic techniques and therefore, in many of those cases, only their protein coding sequences are defined. Some also lack experimental support, due in part to the similarity between them and their monogenic, cell-specific expression in olfactory tissues. Here we use deep RNA sequencing, expression microarray and quantitative RT-PCR in both the vomeronasal organ and whole olfactory mucosa to quantify their full transcriptomes in multiple male and female mice. We find evidence of expression for all VR, and almost all OR genes that are annotated as functional in the reference genome, and use the data to generate over 1100 new, multi-exonic, significantly extended receptor gene annotations. We find that OR and VR genes are neither equally nor randomly expressed, but have reproducible distributions of abundance in both tissues. The olfactory transcriptomes are only minimally different between males and females, suggesting altered gene expression at the periphery is unlikely to underpin the striking sexual dimorphism in olfactory-mediated behavior. Finally, we present evidence that hundreds of novel, putatively protein-coding genes are expressed in these highly specialized olfactory tissues, and carry out a proof-of-principle validation. Taken together, these data provide a comprehensive, quantitative catalog of the genes that mediate olfactory perception and pheromone-evoked behavior at the periphery. The sense of smell in mice involves the detection of odors and pheromones by many hundreds of olfactory and vomeronasal receptors. The genes that encode these receptors account for around 5% of the whole gene catalog, but they are poorly understood because they are very similar to each other, and are thought to be turned on randomly in only a small number of cells. Here we use multiple gene expression technologies to curate and measure the activity of all the genes involved in the detection of odors and find evidence of many new ones. We show that most genes encoding olfactory and vomeronasal receptors have complex, multi-exonic structures that generate different isoforms. We find that some receptors are consistently more abundant in the nose than others, which suggests they are not turned on randomly. This may explain why mice are particularly sensitive to some odors, but less attuned to others. We find that overall males and females differ very little in gene expression, despite having altered behavioral responses to the same odors. Thus diversity in receptor expression can explain differences in odor sensitivity, but does not appear to dictate whether sex pheromones are differentially detected by males or females.
Collapse
|
38
|
Grison A, Zucchelli S, Urzì A, Zamparo I, Lazarevic D, Pascarella G, Roncaglia P, Giorgetti A, Garcia-Esparcia P, Vlachouli C, Simone R, Persichetti F, Forrest ARR, Hayashizaki Y, Carloni P, Ferrer I, Lodovichi C, Plessy C, Carninci P, Gustincich S. Mesencephalic dopaminergic neurons express a repertoire of olfactory receptors and respond to odorant-like molecules. BMC Genomics 2014; 15:729. [PMID: 25164183 PMCID: PMC4161876 DOI: 10.1186/1471-2164-15-729] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 08/18/2014] [Indexed: 01/15/2023] Open
Abstract
Background The mesencephalic dopaminergic (mDA) cell system is composed of two major groups of projecting cells in the Substantia Nigra (SN) (A9 neurons) and the Ventral Tegmental Area (VTA) (A10 cells). Selective degeneration of A9 neurons occurs in Parkinson’s disease (PD) while abnormal function of A10 cells has been linked to schizophrenia, attention deficit and addiction. The molecular basis that underlies selective vulnerability of A9 and A10 neurons is presently unknown. Results By taking advantage of transgenic labeling, laser capture microdissection coupled to nano Cap-Analysis of Gene Expression (nanoCAGE) technology on isolated A9 and A10 cells, we found that a subset of Olfactory Receptors (OR)s is expressed in mDA neurons. Gene expression analysis was integrated with the FANTOM5 Helicos CAGE sequencing datasets, showing the presence of these ORs in selected tissues and brain areas outside of the olfactory epithelium. OR expression in the mesencephalon was validated by RT-PCR and in situ hybridization. By screening 16 potential ligands on 5 mDA ORs recombinantly expressed in an heterologous in vitro system, we identified carvone enantiomers as agonists at Olfr287 and able to evoke an intracellular Ca2+ increase in solitary mDA neurons. ORs were found expressed in human SN and down-regulated in PD post mortem brains. Conclusions Our study indicates that mDA neurons express ORs and respond to odor-like molecules providing new opportunities for pharmacological intervention in disease. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-729) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Piero Carninci
- SISSA, Area of Neuroscience, via Bonomea 265, 34136 Trieste, Italy.
| | | |
Collapse
|
39
|
Li M, Tian S, Yeung CKL, Meng X, Tang Q, Niu L, Wang X, Jin L, Ma J, Long K, Zhou C, Cao Y, Zhu L, Bai L, Tang G, Gu Y, Jiang A, Li X, Li R. Whole-genome sequencing of Berkshire (European native pig) provides insights into its origin and domestication. Sci Rep 2014; 4:4678. [PMID: 24728479 PMCID: PMC3985078 DOI: 10.1038/srep04678] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/28/2014] [Indexed: 01/24/2023] Open
Abstract
Domesticated organisms have experienced strong selective pressures directed at genes or genomic regions controlling traits of biological, agricultural or medical importance. The genome of native and domesticated pigs provide a unique opportunity for tracing the history of domestication and identifying signatures of artificial selection. Here we used whole-genome sequencing to explore the genetic relationships among the European native pig Berkshire and breeds that are distributed worldwide, and to identify genomic footprints left by selection during the domestication of Berkshire. Numerous nonsynonymous SNPs-containing genes fall into olfactory-related categories, which are part of a rapidly evolving superfamily in the mammalian genome. Phylogenetic analyses revealed a deep phylogenetic split between European and Asian pigs rather than between domestic and wild pigs. Admixture analysis exhibited higher portion of Chinese genetic material for the Berkshire pigs, which is consistent with the historical record regarding its origin. Selective sweep analyses revealed strong signatures of selection affecting genomic regions that harbor genes underlying economic traits such as disease resistance, pork yield, fertility, tameness and body length. These discoveries confirmed the history of origin of Berkshire pig by genome-wide analysis and illustrate how domestication has shaped the patterns of genetic variation.
Collapse
Affiliation(s)
- Mingzhou Li
- 1] Biodynamic Optical Imaging Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, and School of Life Sciences, Peking University, Beijing 100871, People's Republic of China [2] Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China [3]
| | - Shilin Tian
- 1] Novogene Bioinformatics Institute, Beijing 100083, People's Republic of China [2]
| | - Carol K L Yeung
- 1] Novogene Bioinformatics Institute, Beijing 100083, People's Republic of China [2]
| | - Xuehong Meng
- Novogene Bioinformatics Institute, Beijing 100083, People's Republic of China
| | - Qianzi Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Lili Niu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Xun Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Long Jin
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Jideng Ma
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Keren Long
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Chaowei Zhou
- 1] Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China [2] Department of Animal Science, Southwest University at Rongchang, Chongqing 402460, People's Republic of China
| | - Yinchuan Cao
- Novogene Bioinformatics Institute, Beijing 100083, People's Republic of China
| | - Li Zhu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Lin Bai
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Guoqing Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Yiren Gu
- Sichuan Animal Science Academy, Chengdu 610066, People's Republic of China
| | - An'an Jiang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Xuewei Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Ruiqiang Li
- 1] Biodynamic Optical Imaging Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, and School of Life Sciences, Peking University, Beijing 100871, People's Republic of China [2] Novogene Bioinformatics Institute, Beijing 100083, People's Republic of China
| |
Collapse
|
40
|
Ignatieva EV, Levitsky VG, Yudin NS, Moshkin MP, Kolchanov NA. Genetic basis of olfactory cognition: extremely high level of DNA sequence polymorphism in promoter regions of the human olfactory receptor genes revealed using the 1000 Genomes Project dataset. Front Psychol 2014; 5:247. [PMID: 24715883 PMCID: PMC3970011 DOI: 10.3389/fpsyg.2014.00247] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 03/05/2014] [Indexed: 11/13/2022] Open
Abstract
The molecular mechanism of olfactory cognition is very complicated. Olfactory cognition is initiated by olfactory receptor proteins (odorant receptors), which are activated by olfactory stimuli (ligands). Olfactory receptors are the initial player in the signal transduction cascade producing a nerve impulse, which is transmitted to the brain. The sensitivity to a particular ligand depends on the expression level of multiple proteins involved in the process of olfactory cognition: olfactory receptor proteins, proteins that participate in signal transduction cascade, etc. The expression level of each gene is controlled by its regulatory regions, and especially, by the promoter [a region of DNA about 100–1000 base pairs long located upstream of the transcription start site (TSS)]. We analyzed single nucleotide polymorphisms using human whole-genome data from the 1000 Genomes Project and revealed an extremely high level of single nucleotide polymorphisms in promoter regions of olfactory receptor genes and HLA genes. We hypothesized that the high level of polymorphisms in olfactory receptor promoters was responsible for the diversity in regulatory mechanisms controlling the expression levels of olfactory receptor proteins. Such diversity of regulatory mechanisms may cause the great variability of olfactory cognition of numerous environmental olfactory stimuli perceived by human beings (air pollutants, human body odors, odors in culinary etc.). In turn, this variability may provide a wide range of emotional and behavioral reactions related to the vast variety of olfactory stimuli.
Collapse
Affiliation(s)
- Elena V Ignatieva
- Laboratory of Evolutionary Bioinformatics and Theoretical Genetics, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences Novosibirsk, Russia ; Department of Natural Science, Novosibirsk State University Novosibirsk, Russia
| | - Victor G Levitsky
- Department of Natural Science, Novosibirsk State University Novosibirsk, Russia ; Laboratory of Molecular-Genetic Systems, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences Novosibirsk, Russia
| | - Nikolay S Yudin
- Department of Natural Science, Novosibirsk State University Novosibirsk, Russia ; Laboratory of Human Molecular Genetics, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences Novosibirsk, Russia
| | - Mikhail P Moshkin
- Department of Natural Science, Novosibirsk State University Novosibirsk, Russia ; Laboratory of Mammalian Ecological Genetics, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences Novosibirsk, Russia
| | - Nikolay A Kolchanov
- Department of Natural Science, Novosibirsk State University Novosibirsk, Russia ; Department of Systems Biology, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences Novosibirsk, Russia ; National Research centre "Kurchatov Institute" Moscow, Russia
| |
Collapse
|
41
|
Irizar H, Muñoz-Culla M, Sepúlveda L, Sáenz-Cuesta M, Prada Á, Castillo-Triviño T, Zamora-López G, de Munain AL, Olascoaga J, Otaegui D. Transcriptomic profile reveals gender-specific molecular mechanisms driving multiple sclerosis progression. PLoS One 2014; 9:e90482. [PMID: 24587374 PMCID: PMC3938749 DOI: 10.1371/journal.pone.0090482] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 01/31/2014] [Indexed: 12/21/2022] Open
Abstract
Background Although the most common clinical presentation of multiple sclerosis (MS) is the so called Relapsing-Remitting MS (RRMS), the molecular mechanisms responsible for its progression are currently unknown. To tackle this problem, a whole-genome gene expression analysis has been performed on RRMS patients. Results The comparative analysis of the Affymetrix Human Gene 1.0 ST microarray data from peripheral blood leucocytes obtained from 25 patients in remission and relapse and 25 healthy subjects has revealed 174 genes altered in both remission and relapse, a high proportion of them showing what we have called “mirror pattern”: they are upregulated in remission and downregulated in relapse or vice versa. The coexpression analysis of these genes has shown that they are organized in three female-specific and one male-specific modules. Conclusions The interpretation of the modules of the coexpression network suggests that Epstein-Barr virus (EBV) reactivation of B cells happens in MS relapses; however, qPCR expression data of the viral genes supports that hypothesis only in female patients, reinforcing the notion that different molecular processes drive disease progression in females and males. Besides, we propose that the “primed” state showed by neutrophils in women is an endogenous control mechanism triggered to keep EBV reactivation under control through vitamin B12 physiology. Finally, our results also point towards an important sex-specific role of non-coding RNA in MS.
Collapse
Affiliation(s)
- Haritz Irizar
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute, Donostia-San Sebastian, Spain
- Spanish Multiple Sclerosis Net (REEM), Barcelona, Spain
| | - Maider Muñoz-Culla
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute, Donostia-San Sebastian, Spain
- Spanish Multiple Sclerosis Net (REEM), Barcelona, Spain
| | - Lucia Sepúlveda
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute, Donostia-San Sebastian, Spain
| | - Matías Sáenz-Cuesta
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute, Donostia-San Sebastian, Spain
- Spanish Multiple Sclerosis Net (REEM), Barcelona, Spain
| | - Álvaro Prada
- Spanish Multiple Sclerosis Net (REEM), Barcelona, Spain
- Hospital Universitario Donostia, Immunology Department, Donostia-San Sebastian, Spain
| | - Tamara Castillo-Triviño
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute, Donostia-San Sebastian, Spain
- Spanish Multiple Sclerosis Net (REEM), Barcelona, Spain
| | - Gorka Zamora-López
- Bernstein Center for Computational Neuroscience, Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Brain and Cognition, Universistat Pompeu Fabra, Barcelona, Spain
| | - Adolfo López de Munain
- Hospital Universitario Donostia, Neurology Department, Donostia-San Sebastian, Spain
- Centro de Investigaciones Biomédicas en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, Ministerio de Ciencia e Innovación, Madrid, Spain
| | - Javier Olascoaga
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute, Donostia-San Sebastian, Spain
- Spanish Multiple Sclerosis Net (REEM), Barcelona, Spain
- Hospital Universitario Donostia, Neurology Department, Donostia-San Sebastian, Spain
| | - David Otaegui
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute, Donostia-San Sebastian, Spain
- Spanish Multiple Sclerosis Net (REEM), Barcelona, Spain
- * E-mail:
| |
Collapse
|
42
|
Pascarella G, Lazarevic D, Plessy C, Bertin N, Akalin A, Vlachouli C, Simone R, Faulkner GJ, Zucchelli S, Kawai J, Daub CO, Hayashizaki Y, Lenhard B, Carninci P, Gustincich S. NanoCAGE analysis of the mouse olfactory epithelium identifies the expression of vomeronasal receptors and of proximal LINE elements. Front Cell Neurosci 2014; 8:41. [PMID: 24600346 PMCID: PMC3927265 DOI: 10.3389/fncel.2014.00041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/28/2014] [Indexed: 11/13/2022] Open
Abstract
By coupling laser capture microdissection to nanoCAGE technology and next-generation sequencing we have identified the genome-wide collection of active promoters in the mouse Main Olfactory Epithelium (MOE). Transcription start sites (TSSs) for the large majority of Olfactory Receptors (ORs) have been previously mapped increasing our understanding of their promoter architecture. Here we show that in our nanoCAGE libraries of the mouse MOE we detect a large number of tags mapped in loci hosting Type-1 and Type-2 Vomeronasal Receptors genes (V1Rs and V2Rs). These loci also show a massive expression of Long Interspersed Nuclear Elements (LINEs). We have validated the expression of selected receptors detected by nanoCAGE with in situ hybridization, RT-PCR and qRT-PCR. This work extends the repertory of receptors capable of sensing chemical signals in the MOE, suggesting intriguing interplays between MOE and VNO for pheromone processing and positioning transcribed LINEs as candidate regulatory RNAs for VRs expression.
Collapse
Affiliation(s)
- Giovanni Pascarella
- Area of Neuroscience, International School for Advanced Studies (SISSA) Trieste, Italy ; RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Dejan Lazarevic
- Area of Neuroscience, International School for Advanced Studies (SISSA) Trieste, Italy ; Cluster in Biomedicine (CBM), AREA Science Park Trieste, Italy
| | - Charles Plessy
- RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Nicolas Bertin
- RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Altuna Akalin
- Bergen Center for Computational Science - Computational Biology Unit and Sars Centre for Marine Molecular Biology, University of Bergen Bergen, Norway
| | - Christina Vlachouli
- Area of Neuroscience, International School for Advanced Studies (SISSA) Trieste, Italy
| | - Roberto Simone
- Area of Neuroscience, International School for Advanced Studies (SISSA) Trieste, Italy
| | - Geoffrey J Faulkner
- Cancer Biology Program, Mater Medical Research Institute South Brisbane, QLD, Australia ; School of Biomedical Sciences, University of Queensland Brisbane, QLD, Australia
| | - Silvia Zucchelli
- Area of Neuroscience, International School for Advanced Studies (SISSA) Trieste, Italy ; Department of Health Sciences, University of Eastern Piedmont "A. Avogadro," Novara, Italy
| | - Jun Kawai
- RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Carsten O Daub
- RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Yoshihide Hayashizaki
- RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Boris Lenhard
- Bergen Center for Computational Science - Computational Biology Unit and Sars Centre for Marine Molecular Biology, University of Bergen Bergen, Norway
| | - Piero Carninci
- RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Stefano Gustincich
- Area of Neuroscience, International School for Advanced Studies (SISSA) Trieste, Italy
| |
Collapse
|
43
|
Abstract
Odorants are detected by odorant receptors, which are located on olfactory sensory neurons of the nose. Each olfactory sensory neuron expresses one single odorant receptor gene allele from a large family of odorant receptor genes. To gain insight into the mechanisms underlying this monogenic and monoallelic expression, we examined the 3D nuclear organization of olfactory sensory neurons and determined the positions of homologous odorant receptor gene alleles in relation to different nuclear compartments. Our results show that olfactory neurons exhibit a singular nuclear architecture that is characterized by a large centrally localized constitutive heterochromatin block and by the presence of prominent facultative heterochromatin domains that are localized around this constitutive heterochromatin block. We also found that the two homologous alleles of a given odorant receptor gene are frequently segregated to separate compartments in the nucleus, with one of the alleles localized to the constitutive heterochromatin block and the other one localized to the more plastic facultative heterochromatin, or next to it. Our findings suggest that this nuclear compartmentalization may play a critical role in the expression of odorant receptor genes.
Collapse
|
44
|
Fonseca-Sanchéz MA, Pérez-Plasencia C, Fernández-Retana J, Arechaga-Ocampo E, Marchat LA, Rodríguez-Cuevas S, Bautista-Piña V, Arellano-Anaya ZE, Flores-Pérez A, Diaz-Chávez J, López-Camarillo C. microRNA-18b is upregulated in breast cancer and modulates genes involved in cell migration. Oncol Rep 2013; 30:2399-2410. [PMID: 23970382 DOI: 10.3892/or.2013.2691] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/02/2013] [Indexed: 11/06/2022] Open
Abstract
microRNAs are small non-coding RNAs of ~22 nucleotides that function at post-transcriptional level as negative regulators of gene expression. Aberrant expression of microRNAs could promote uncontrolled proliferation, migration and invasion of human cancer cells. In this study, we analyzed the expression of microRNA-18b (miR-18b) in breast cancer cell lines and in a set of clinical specimens. Our results showed that miR-18b was upregulated in four out of five breast cancer cell lines and also in breast tumors. In order to identify potential gene targets, we carried out transcriptional profiling of MDA-MB-231 breast cancer cells that ectopically expressed miR-18b. Our results showed that 263 genes were significantly modulated in miR-18b-deficient cells (fold change >1.5; P≤0.05). We found that knock-down of miR-18b induced the upregulation of 55 olfactory receptor (OR) genes and nine genes (NLRP7, KLK3, OLFM3, POSTN, MAGED4B, KIR3DL3, CRX, SEMG1 and CEACAM5) with key roles in cell migration and metastasis. Consistently, we found that ectopic inhibition of miR-18b suppressed the migration of two breast cancer cell models in vitro. In conclusion, we have uncovered genes directly or indirectly modulated by miR-18b which may represent potential therapeutic targets in breast cancer. Our data also pointed out a role of miR-18b in migration of breast cancer cells.
Collapse
Affiliation(s)
- Miguel A Fonseca-Sanchéz
- Oncogenomics and Cancer Proteomics Laboratory, Genomics Sciences Program, Autonomous University of Mexico City, Mexico City, Mexico
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Strbenac D, Armstrong NJ, Yang JYH. Detection and classification of peaks in 5' cap RNA sequencing data. BMC Genomics 2013; 14 Suppl 5:S9. [PMID: 24564843 PMCID: PMC3852351 DOI: 10.1186/1471-2164-14-s5-s9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The large-scale sequencing of 5' cap enriched cDNA promises to reveal the diversity of transcription initiation across entire genomes. The process of transcription is noisy, and there is often no single, exact start site. This creates the need for a fast and simple method of identifying transcription start peaks based on this type of data. Due to both biological and technical noise, many of the peaks seen are not real transcription initiation events. Classification of the observed peaks is an essential filtering step in the discovery of genuine initiation locations. RESULTS We develop a two-stage approach consisting of a fast and simple algorithm based on a sliding window with Poisson null distribution for detecting the genomic locations of peaks, followed by a linear support vector machine classifier to distinguish between peaks which represent the initiation of transcription and peaks that do not. Comparison of classification performance to the best existing method based on whole genome segmentation showed comparable precision and improved recall. Internal features, which are intrinsic to the data and require no further experiments, had high precision and recall rates. Addition of pooled external data or matched RNA sequencing data resulted in gains of recall with equivalent precision. CONCLUSIONS The Poisson sliding window model is an effective and fast way of taking the peak neighbourhood into account, and finding statistically significant peaks over a range of transcript expression values. It is orders of magnitude faster than doing whole genome segmentation. The support vector classification scheme has better precision and recall than existing methods. Integrating additional datasets is shown to provide minor gains in recall, in comparison to using only the cap-sequencing data.
Collapse
|
46
|
Engineered mitochondrial ferritin as a magnetic resonance imaging reporter in mouse olfactory epithelium. PLoS One 2013; 8:e72720. [PMID: 24023635 PMCID: PMC3758330 DOI: 10.1371/journal.pone.0072720] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 07/12/2013] [Indexed: 12/16/2022] Open
Abstract
We report the design of a MRI reporter gene with applications to non-invasive molecular imaging. We modified mitochondrial ferritin to localize to the cell cytoplasm. We confirmed the efficient cellular processing of this engineered protein and demonstrated high iron loading in mammalian cells. The reporter’s intracellular localization appears as distinct clusters that deliver robust MRI contrast. We used this new reporter to image in vivo and ex vivo the gene expression in native olfactory sensory neurons in the mouse epithelium. This robust MRI reporter can facilitate the study of the molecular mechanisms of olfaction and to monitor intranasal gene therapy delivery, as well as a wide range of cell tracking and gene expression studies in living subjects.
Collapse
|
47
|
Fleischmann A, Abdus-Saboor I, Sayed A, Shykind B. Functional interrogation of an odorant receptor locus reveals multiple axes of transcriptional regulation. PLoS Biol 2013; 11:e1001568. [PMID: 23700388 PMCID: PMC3660300 DOI: 10.1371/journal.pbio.1001568] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 04/12/2013] [Indexed: 11/30/2022] Open
Abstract
A transgenic approach in mice allows the functional interrogation of an odorant receptor locus in vivo and reveals characteristics of its monogenic and monoallelic expression. The odorant receptor (OR) genes constitute the largest mammalian gene family and are expressed in a monogenic and monoallelic fashion, through an unknown mechanism that likely exploits positive and negative regulation. We devised a genetic strategy in mice to examine OR selection by determining the transcriptional activity of an exogenous promoter homologously integrated into an OR locus. Using the tetracycline-dependent transactivator responsive promoter (teto), we observed that the OR locus imposes spatial and temporal constraints on teto-driven transcription. Conditional expression experiments reveal a developmental change in the permissiveness of the locus. Further, expression of an OR transgene that suppresses endogenous ORs similarly represses the OR-integrated teto. Neurons homozygous for the teto-modified allele demonstrate predominantly monoallelic expression, despite their potential to express both copies. These data reveal multiple axes of regulation, and support a model of initiation of OR choice limited by nonpermissive chromatin and maintained by repression of nonselected alleles. Odorant receptor (OR) gene choice is a paradigmatic example of transcriptional regulation in which each olfactory sensory neuron selects a single OR from a repertoire of over 1,000 genes. Two mechanistic models of OR choice have been proposed. One postulates the existence of a specialized transcriptional machinery that selects just one OR allele, while a second, kinetic model proposes that OR chromatin is intrinsically nonpermissive, such that inefficient activation during a critical window of time restricts expression to a single OR allele. Here, we used a transgenic approach in mice in which we inserted a conditionally regulated exogenous promoter into an OR locus by homologous recombination in embryonic stem cells. The resulting novel mouse lines allowed the functional interrogation of the OR locus in vivo during development of the olfactory epithelium, enabling us to directly test models of OR choice. Using this experimental strategy we found that OR loci are indeed slow to activate and that the subsequent phenomenon of spatial restriction of OR expression is accomplished by repression. We also observed a developmental shutdown of OR loci concomitant with expression of the OR repertoire. Together, these experiments provide prima facie evidence for a kinetic model of initiation of OR gene choice, coupled with repression of nonselected OR alleles.
Collapse
Affiliation(s)
| | - Ishmail Abdus-Saboor
- Weill Cornell Medical College in Qatar, Qatar Foundation–Education City, Doha, Qatar
| | - Atef Sayed
- Weill Cornell Medical College in Qatar, Qatar Foundation–Education City, Doha, Qatar
| | - Benjamin Shykind
- Weill Cornell Medical College in Qatar, Qatar Foundation–Education City, Doha, Qatar
- * E-mail:
| |
Collapse
|
48
|
Metazoan promoters: emerging characteristics and insights into transcriptional regulation. Nat Rev Genet 2012; 13:233-45. [PMID: 22392219 DOI: 10.1038/nrg3163] [Citation(s) in RCA: 358] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Promoters are crucial for gene regulation. They vary greatly in terms of associated regulatory elements, sequence motifs, the choice of transcription start sites and other features. Several technologies that harness next-generation sequencing have enabled recent advances in identifying promoters and their features, helping researchers who are investigating functional categories of promoters and their modes of regulation. Additional features of promoters that are being characterized include types of histone modifications, nucleosome positioning, RNA polymerase pausing and novel small RNAs. In this Review, we discuss recent findings relating to metazoan promoters and how these findings are leading to a revised picture of what a gene promoter is and how it works.
Collapse
|