1
|
Saab C, Stephan J, Akoury E. Structural insights into the binding mechanism of Clr4 methyltransferase to H3K9 methylated nucleosome. Sci Rep 2024; 14:5438. [PMID: 38443490 PMCID: PMC10914790 DOI: 10.1038/s41598-024-56248-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/04/2024] [Indexed: 03/07/2024] Open
Abstract
The establishment and maintenance of heterochromatin, a specific chromatin structure essential for genomic stability and regulation, rely on intricate interactions between chromatin-modifying enzymes and nucleosomal histone proteins. However, the precise trigger for these modifications remains unclear, thus highlighting the need for a deeper understanding of how methyltransferases facilitate histone methylation among others. Here, we investigate the molecular mechanisms underlying heterochromatin assembly by studying the interaction between the H3K9 methyltransferase Clr4 and H3K9-methylated nucleosomes. Using a combination of liquid-state nuclear magnetic resonance spectroscopy and cryo-electron microscopy, we elucidate the structural basis of Clr4 binding to H3K9-methylated nucleosomes. Our results reveal that Clr4 engages with nucleosomes through its chromodomain and disordered regions to promote de novo methylation. This study provides crucial insights into the molecular mechanisms governing heterochromatin formation by highlighting the significance of chromatin-modifying enzymes in genome regulation and disease pathology.
Collapse
Affiliation(s)
- Christopher Saab
- Department of Natural Sciences, Lebanese American University, Beirut, 1102-2801, Lebanon
- Department of Chemistry, McGill University, 801 Sherbrooke St. West, Montreal, QC, H3AOB8, Canada
| | - Joseph Stephan
- School of Medicine, Lebanese American University, PO Box 36, Byblos, Lebanon
| | - Elias Akoury
- Department of Natural Sciences, Lebanese American University, Beirut, 1102-2801, Lebanon.
| |
Collapse
|
2
|
Banu MSA, Huda KMK, Harun-Ur-Rashid M, Parveen S, Tuteja N. A DEAD box helicase Psp68 positively regulates salt stress responses in marker-free transgenic rice plants. Transgenic Res 2023; 32:293-304. [PMID: 37247124 DOI: 10.1007/s11248-023-00353-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/16/2023] [Indexed: 05/30/2023]
Abstract
Helicases are the motor proteins not only involved in transcriptional and post-transcription process but also provide abiotic stress tolerance in many crops. The p68, belong to the SF2 (DEAD-box helicase) family proteins and overexpression of Psp68 providing enhanced tolerance to transgenic rice plants. In this study, salinity tolerant marker-free transgenic rice has been developed by overexpressing Psp68 gene and phenotypically characterized. The Psp68 overexpressing marker-free transgenic rice plants were initially screened in the rooting medium containing salt stress and 20% polyethylene glycol (PEG). Stable integration and overexpression of Psp68 in marker-free transgenic lines were confirmed by molecular analyses including PCR, southern, western blot, and qRT-PCR analyses. The marker-free transgenic lines showed enhanced tolerance to salinity stress as displayed by early seed germination, higher chlorophyll content, reduced necrosis, more survival rate, improved seedling growth and more grain yield per plant. Furthermore, Psp68 overexpressing marker-free transgenics also accumulated less Na+ and higher K+ ions in the presence of salinity stress. Phenotypic analyses also revealed that marker-free transgenic rice lines efficiently scavenge ROS-mediated damages as displayed by lower H2O2 and malondialdehyde content, delayed electrolyte leakage, higher photosynthetic efficiency, membrane stability, proline content and enhanced activities of antioxidants enzymes. Overall, our results confirmed that Psp68 overexpression confers salinity stress tolerance in marker-free transgenics, hence the technique could be utilized to develop genetically modified crops without any biosafety issues.
Collapse
Affiliation(s)
- Mst Sufara Akhter Banu
- Bangladesh Agricultural Research Council (BARC), Dhaka, 1215, Bangladesh
- Plant Molecular Biology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi, Delhi, 110067, India
| | - Kazi Md Kamrul Huda
- Department of Genetics and Plant Breeding, Sher-e-Bangla Agricultural University, Dhaka, 1207, Bangladesh.
- Plant Molecular Biology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi, Delhi, 110067, India.
| | - Md Harun-Ur-Rashid
- Department of Genetics and Plant Breeding, Sher-e-Bangla Agricultural University, Dhaka, 1207, Bangladesh
| | - Shahanaz Parveen
- Department of Genetics and Plant Breeding, Sher-e-Bangla Agricultural University, Dhaka, 1207, Bangladesh
| | - Narendra Tuteja
- Plant Molecular Biology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi, Delhi, 110067, India
| |
Collapse
|
3
|
Fingerhut JM, Yamashita YM. The regulation and potential functions of intronic satellite DNA. Semin Cell Dev Biol 2022; 128:69-77. [PMID: 35469677 DOI: 10.1016/j.semcdb.2022.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022]
Abstract
Satellite DNAs are arrays of tandem repeats found in the eukaryotic genome. They are mainly found in pericentromeric heterochromatin and have been believed to be mostly inert, leading satellite DNAs to be erroneously regarded as junk. Recent studies have started to elucidate the function of satellite DNA, yet little is known about the peculiar case where satellite DNA is found within the introns of protein coding genes, resulting in incredibly large introns, a phenomenon termed intron gigantism. Studies in Drosophila demonstrated that satellite DNA-containing introns are transcribed with the gene and require specialized mechanisms to overcome the burdens imposed by the extremely long stretches of repetitive DNA. Whether intron gigantism confers any benefit or serves any functional purpose for cells and/or organisms remains elusive. Here we review our current understanding of intron gigantism: where it is found, the challenges it imposes, how it is regulated and what purpose it may serve.
Collapse
Affiliation(s)
- Jaclyn M Fingerhut
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Howard Hughes Medical Institute, Cambridge, MA, USA.
| | - Yukiko M Yamashita
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Howard Hughes Medical Institute, Cambridge, MA, USA.
| |
Collapse
|
4
|
Bughio FJ, Maggert KA. Live analysis of position-effect variegation in Drosophila reveals different modes of action for HP1a and Su(var)3-9. Proc Natl Acad Sci U S A 2022; 119:e2118796119. [PMID: 35704756 PMCID: PMC9231485 DOI: 10.1073/pnas.2118796119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 04/19/2022] [Indexed: 11/18/2022] Open
Abstract
Position-effect variegation (PEV) results from the juxtaposition of euchromatic and heterochromatic components of eukaryotic genomes, silencing genes near the new euchromatin/heterochromatin junctions. Silencing is itself heritable through S phase, giving rise to distinctive random patterns of cell clones expressing the genes intermixed with clones in which the genes are silenced. Much of what we know about epigenetic inheritance in the soma stems from work on PEV aimed at identifying the components of the silencing machinery and its mechanism of inheritance. The roles of two central gene activities-the Su(var)3-9-encoded histone H3-lysine-9 methyltransferase and the Su(var)205-encoded methyl-H3-lysine-9 binding protein heterochromatin protein 1 (HP1a)-have been inferred from terminal phenotypes, leaving considerable gaps in understanding of how PEV behaves through development. Here, we investigate the PEV phenotypes of Su(var)3-9 and Su(var)205 mutations in live developing tissues. We discovered that mutation in Su(var)205 compromises the initial establishment of PEV in early embryogenesis. Later gains of heterochromatin-induced gene silencing are possible but are unstable and lost rapidly. In contrast, a strain with mutation in Su(var)3-9 exhibits robust silencing early in development but fails to maintain it through subsequent cell divisions. Our analyses show that, while the terminal phenotypes of these mutations may appear identical, they have arrived at them through different developmental trajectories. We discuss how our findings expand and clarify existing models for epigenetic inheritance of heterochromatin-induced gene silencing.
Collapse
Affiliation(s)
- Farah J. Bughio
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | - Keith A. Maggert
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| |
Collapse
|
5
|
Cotsworth S, Jackson CJ, Hallson G, Fitzpatrick KA, Syrzycka M, Coulthard AB, Bejsovec A, Marchetti M, Pimpinelli S, Wang SJH, Camfield RG, Verheyen EM, Sinclair DA, Honda BM, Hilliker AJ. Characterization of Gfat1 ( zeppelin) and Gfat2, Essential Paralogous Genes Which Encode the Enzymes That Catalyze the Rate-Limiting Step in the Hexosamine Biosynthetic Pathway in Drosophila melanogaster. Cells 2022; 11:448. [PMID: 35159258 PMCID: PMC8834284 DOI: 10.3390/cells11030448] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 11/16/2022] Open
Abstract
The zeppelin (zep) locus is known for its essential role in the development of the embryonic cuticle of Drosophila melanogaster. We show here that zep encodes Gfat1 (Glutamine: Fructose-6-Phosphate Aminotransferase 1; CG12449), the enzyme that catalyzes the rate-limiting step in the hexosamine biosynthesis pathway (HBP). This conserved pathway diverts 2%-5% of cellular glucose from glycolysis and is a nexus of sugar (fructose-6-phosphate), amino acid (glutamine), fatty acid [acetyl-coenzymeA (CoA)], and nucleotide/energy (UDP) metabolism. We also describe the isolation and characterization of lethal mutants in the euchromatic paralog, Gfat2 (CG1345), and demonstrate that ubiquitous expression of Gfat1+ or Gfat2+ transgenes can rescue lethal mutations in either gene. Gfat1 and Gfat2 show differences in mRNA and protein expression during embryogenesis and in essential tissue-specific requirements for Gfat1 and Gfat2, suggesting a degree of functional evolutionary divergence. An evolutionary, cytogenetic analysis of the two genes in six Drosophila species revealed Gfat2 to be located within euchromatin in all six species. Gfat1 localizes to heterochromatin in three melanogaster-group species, and to euchromatin in the more distantly related species. We have also found that the pattern of flanking-gene microsynteny is highly conserved for Gfat1 and somewhat less conserved for Gfat2.
Collapse
Affiliation(s)
- Shawn Cotsworth
- Department of Molecular Biology and Biochemistry (MBB), Simon Fraser University, 8888 University Dr., Burnaby, BC V5A 1S6, Canada; (S.C.); (C.J.J.); (G.H.); (K.A.F.); (M.S.); (S.J.H.W.); (E.M.V.); (D.A.S.); (B.M.H.)
| | - Catherine J. Jackson
- Department of Molecular Biology and Biochemistry (MBB), Simon Fraser University, 8888 University Dr., Burnaby, BC V5A 1S6, Canada; (S.C.); (C.J.J.); (G.H.); (K.A.F.); (M.S.); (S.J.H.W.); (E.M.V.); (D.A.S.); (B.M.H.)
- Department of Plastic and Reconstructive Surgery, Institute for Surgical Research, University of Oslo, N-0424 Oslo, Norway
- The Department of Medical Biochemistry, Oslo University Hospital, N-0424 Oslo, Norway
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, N-0424 Oslo, Norway
| | - Graham Hallson
- Department of Molecular Biology and Biochemistry (MBB), Simon Fraser University, 8888 University Dr., Burnaby, BC V5A 1S6, Canada; (S.C.); (C.J.J.); (G.H.); (K.A.F.); (M.S.); (S.J.H.W.); (E.M.V.); (D.A.S.); (B.M.H.)
| | - Kathleen A. Fitzpatrick
- Department of Molecular Biology and Biochemistry (MBB), Simon Fraser University, 8888 University Dr., Burnaby, BC V5A 1S6, Canada; (S.C.); (C.J.J.); (G.H.); (K.A.F.); (M.S.); (S.J.H.W.); (E.M.V.); (D.A.S.); (B.M.H.)
| | - Monika Syrzycka
- Department of Molecular Biology and Biochemistry (MBB), Simon Fraser University, 8888 University Dr., Burnaby, BC V5A 1S6, Canada; (S.C.); (C.J.J.); (G.H.); (K.A.F.); (M.S.); (S.J.H.W.); (E.M.V.); (D.A.S.); (B.M.H.)
- Allergan Canada, 500-85 Enterprise Blvd, Markham, ON L6G 0B5, Canada
| | | | - Amy Bejsovec
- Department of Biology, Duke University, Durham, NC 27708, USA;
| | - Marcella Marchetti
- Department of Biology and Biotechnology “C. Darwin”, “Sapienza” University of Rome, 00185 Rome, Italy; (M.M.); (S.P.)
| | - Sergio Pimpinelli
- Department of Biology and Biotechnology “C. Darwin”, “Sapienza” University of Rome, 00185 Rome, Italy; (M.M.); (S.P.)
| | - Simon J. H. Wang
- Department of Molecular Biology and Biochemistry (MBB), Simon Fraser University, 8888 University Dr., Burnaby, BC V5A 1S6, Canada; (S.C.); (C.J.J.); (G.H.); (K.A.F.); (M.S.); (S.J.H.W.); (E.M.V.); (D.A.S.); (B.M.H.)
| | - Robert G. Camfield
- BC Genome Science Centre, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada;
| | - Esther M. Verheyen
- Department of Molecular Biology and Biochemistry (MBB), Simon Fraser University, 8888 University Dr., Burnaby, BC V5A 1S6, Canada; (S.C.); (C.J.J.); (G.H.); (K.A.F.); (M.S.); (S.J.H.W.); (E.M.V.); (D.A.S.); (B.M.H.)
| | - Donald A. Sinclair
- Department of Molecular Biology and Biochemistry (MBB), Simon Fraser University, 8888 University Dr., Burnaby, BC V5A 1S6, Canada; (S.C.); (C.J.J.); (G.H.); (K.A.F.); (M.S.); (S.J.H.W.); (E.M.V.); (D.A.S.); (B.M.H.)
| | - Barry M. Honda
- Department of Molecular Biology and Biochemistry (MBB), Simon Fraser University, 8888 University Dr., Burnaby, BC V5A 1S6, Canada; (S.C.); (C.J.J.); (G.H.); (K.A.F.); (M.S.); (S.J.H.W.); (E.M.V.); (D.A.S.); (B.M.H.)
| | | |
Collapse
|
6
|
Zykova T, Maltseva M, Goncharov F, Boldyreva L, Pokholkova G, Kolesnikova T, Zhimulev I. The Organization of Pericentromeric Heterochromatin in Polytene Chromosome 3 of the Drosophilamelanogaster Line with the Rif11; SuURES Su(var)3-906 Mutations Suppressing Underreplication. Cells 2021; 10:2809. [PMID: 34831030 PMCID: PMC8616060 DOI: 10.3390/cells10112809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/11/2021] [Accepted: 10/14/2021] [Indexed: 11/17/2022] Open
Abstract
Although heterochromatin makes up 40% of the Drosophila melanogaster genome, its organization remains little explored, especially in polytene chromosomes, as it is virtually not represented in them due to underreplication. Two all-new approaches were used in this work: (i) with the use of a newly synthesized Drosophila line that carries three mutations, Rif11, SuURES and Su(var)3-906, suppressing the underreplication of heterochromatic regions, we obtained their fullest representation in polytene chromosomes and described their structure; (ii) 20 DNA fragments with known positions on the physical map as well as molecular genetic features of the genome (gene density, histone marks, heterochromatin proteins, origin recognition complex proteins, replication timing sites and satellite DNAs) were mapped in the newly polytenized heterochromatin using FISH and bioinformatics data. The borders of the heterochromatic regions and variations in their positions on arm 3L have been determined for the first time. The newly polytenized heterochromatic material exhibits two main types of morphology: a banding pattern (locations of genes and short satellites) and reticular chromatin (locations of large blocks of satellite DNA). The locations of the banding and reticular polytene heterochromatin was determined on the physical map.
Collapse
Affiliation(s)
- Tatyana Zykova
- Laboratory of Molecular Cytogenetics, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia; (T.Z.); (M.M.); (F.G.); (L.B.); (G.P.); (T.K.)
| | - Mariya Maltseva
- Laboratory of Molecular Cytogenetics, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia; (T.Z.); (M.M.); (F.G.); (L.B.); (G.P.); (T.K.)
| | - Fedor Goncharov
- Laboratory of Molecular Cytogenetics, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia; (T.Z.); (M.M.); (F.G.); (L.B.); (G.P.); (T.K.)
| | - Lidia Boldyreva
- Laboratory of Molecular Cytogenetics, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia; (T.Z.); (M.M.); (F.G.); (L.B.); (G.P.); (T.K.)
| | - Galina Pokholkova
- Laboratory of Molecular Cytogenetics, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia; (T.Z.); (M.M.); (F.G.); (L.B.); (G.P.); (T.K.)
| | - Tatyana Kolesnikova
- Laboratory of Molecular Cytogenetics, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia; (T.Z.); (M.M.); (F.G.); (L.B.); (G.P.); (T.K.)
- Laboratory of Structural, Functional and Comparative Genomics Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Igor Zhimulev
- Laboratory of Molecular Cytogenetics, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia; (T.Z.); (M.M.); (F.G.); (L.B.); (G.P.); (T.K.)
- Laboratory of Structural, Functional and Comparative Genomics Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
7
|
Nguyen AH, Bachtrog D. Toxic Y chromosome: Increased repeat expression and age-associated heterochromatin loss in male Drosophila with a young Y chromosome. PLoS Genet 2021; 17:e1009438. [PMID: 33886541 PMCID: PMC8061872 DOI: 10.1371/journal.pgen.1009438] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
Sex-specific differences in lifespan are prevalent across the tree of life and influenced by heteromorphic sex chromosomes. In species with XY sex chromosomes, females often outlive males. Males and females can differ in their overall repeat content due to the repetitive Y chromosome, and repeats on the Y might lower survival of the heterogametic sex (toxic Y effect). Here, we take advantage of the well-assembled young Y chromosome of Drosophila miranda to study the sex-specific dynamics of chromatin structure and repeat expression during aging in male and female flies. Male D. miranda have about twice as much repetitive DNA compared to females, and live shorter than females. Heterochromatin is crucial for silencing of repetitive elements, yet old D. miranda flies lose H3K9me3 modifications in their pericentromere, with heterochromatin loss being more severe during aging in males than females. Satellite DNA becomes de-repressed more rapidly in old vs. young male flies relative to females. In contrast to what is observed in D. melanogaster, we find that transposable elements (TEs) are expressed at higher levels in male D. miranda throughout their life. We show that epigenetic silencing via heterochromatin formation is ineffective on the TE-rich neo-Y chromosome, presumably due to active transcription of a large number of neo-Y linked genes, resulting in up-regulation of Y-linked TEs already in young males. This is consistent with an interaction between the evolutionary age of the Y chromosome and the genomic effects of aging. Our data support growing evidence that "toxic Y chromosomes" can diminish male fitness and a reduction in heterochromatin can contribute to sex-specific aging.
Collapse
Affiliation(s)
- Alison H. Nguyen
- Department of Integrative Biology, University of California Berkeley, Berkeley, California, United States of America
| | - Doris Bachtrog
- Department of Integrative Biology, University of California Berkeley, Berkeley, California, United States of America
| |
Collapse
|
8
|
Palazzo A, Marsano RM. Transposable elements: a jump toward the future of expression vectors. Crit Rev Biotechnol 2021; 41:792-808. [PMID: 33622117 DOI: 10.1080/07388551.2021.1888067] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Expression vectors (EVs) are artificial nucleic acid molecules with a modular structure that allows for the transcription of DNA sequences of interest in either cellular or cell-free environments. These vectors have emerged as cross-disciplinary tools with multiple applications in an expanding Life Sciences market. The cis-regulatory sequences (CRSs) that control the transcription in EVs are typically sourced from either viruses or from characterized genes. However, the recent advancement in transposable elements (TEs) technology provides attractive alternatives that may enable a significant improvement in the design of EVs. Commonly known as "jumping genes," due to their ability to move between genetic loci, TEs are constitutive components of both eukaryotic and prokaryotic genomes. TEs harbor native CRSs that allow the regulated transcription of transposition-related genes. However, some TE-related CRSs display striking characteristics, which provides the opportunity to reconsider TEs as lead actors in the design of EVs. In this article, we provide a synopsis of the transcriptional control elements commonly found in EVs together with an extensive discussion of their advantages and limitations. We also highlight the latest findings that may allow for the implementation of TE-derived sequences in the EVs feasible, possibly improving existing vectors. By introducing this new concept of TEs as a source of regulatory sequences, we aim to stimulate a profitable discussion of the potential advantages and benefits of developing a new generation of EVs based on the use of TE-derived control sequences.
Collapse
Affiliation(s)
- Antonio Palazzo
- Laboratory of Translational Nanotechnology, "Istituto Tumori Giovanni Paolo II" I.R.C.C.S, Bari, Italy
| | | |
Collapse
|
9
|
Evolutionary Dynamics of the Pericentromeric Heterochromatin in Drosophila virilis and Related Species. Genes (Basel) 2021; 12:genes12020175. [PMID: 33513919 PMCID: PMC7911463 DOI: 10.3390/genes12020175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/21/2021] [Accepted: 01/23/2021] [Indexed: 12/19/2022] Open
Abstract
Pericentromeric heterochromatin in Drosophila generally consists of repetitive DNA, forming the environment associated with gene silencing. Despite the expanding knowledge of the impact of transposable elements (TEs) on the host genome, little is known about the evolution of pericentromeric heterochromatin, its structural composition, and age. During the evolution of the Drosophilidae, hundreds of genes have become embedded within pericentromeric regions yet retained activity. We investigated a pericentromeric heterochromatin fragment found in D. virilis and related species, describing the evolution of genes in this region and the age of TE invasion. Regardless of the heterochromatic environment, the amino acid composition of the genes is under purifying selection. However, the selective pressure affects parts of genes in varying degrees, resulting in expansion of gene introns due to TEs invasion. According to the divergence of TEs, the pericentromeric heterochromatin of the species of virilis group began to form more than 20 million years ago by invasions of retroelements, miniature inverted repeat transposable elements (MITEs), and Helitrons. Importantly, invasions into the heterochromatin continue to occur by TEs that fall under the scope of piRNA silencing. Thus, the pericentromeric heterochromatin, in spite of its ability to induce silencing, has the means for being dynamic, incorporating the regions of active transcription.
Collapse
|
10
|
Kasinathan B, Colmenares SU, McConnell H, Young JM, Karpen GH, Malik HS. Innovation of heterochromatin functions drives rapid evolution of essential ZAD-ZNF genes in Drosophila. eLife 2020; 9:e63368. [PMID: 33169670 PMCID: PMC7655104 DOI: 10.7554/elife.63368] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Contrary to dogma, evolutionarily young and dynamic genes can encode essential functions. We find that evolutionarily dynamic ZAD-ZNF genes, which encode the most abundant class of insect transcription factors, are more likely to encode essential functions in Drosophila melanogaster than ancient, conserved ZAD-ZNF genes. We focus on the Nicknack ZAD-ZNF gene, which is evolutionarily young, poorly retained in Drosophila species, and evolves under strong positive selection. Yet we find that it is necessary for larval development in D. melanogaster. We show that Nicknack encodes a heterochromatin-localizing protein like its paralog Oddjob, also an evolutionarily dynamic yet essential ZAD-ZNF gene. We find that the divergent D. simulans Nicknack protein can still localize to D. melanogaster heterochromatin and rescue viability of female but not male Nicknack-null D. melanogaster. Our findings suggest that innovation for rapidly changing heterochromatin functions might generally explain the essentiality of many evolutionarily dynamic ZAD-ZNF genes in insects.
Collapse
Affiliation(s)
- Bhavatharini Kasinathan
- Medical Scientist Training Program, University of Washington School of MedicineSeattleUnited States
- Molecular and Cellular Biology Graduate program, University of Washington School of MedicineSeattleUnited States
- Division of Basic Sciences, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Serafin U Colmenares
- Biological Systems and Engineering Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
- Department of Molecular and Cell Biology, University of California at BerkeleyBerkeleyUnited States
- Innovative Genomics InstituteBerkeleyUnited States
| | - Hannah McConnell
- Division of Basic Sciences, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Janet M Young
- Division of Basic Sciences, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Gary H Karpen
- Biological Systems and Engineering Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
- Department of Molecular and Cell Biology, University of California at BerkeleyBerkeleyUnited States
- Innovative Genomics InstituteBerkeleyUnited States
| | - Harmit S Malik
- Division of Basic Sciences, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Howard Hughes Medical Institute, Fred Hutchinson Cancer Research CenterSeattleUnited States
| |
Collapse
|
11
|
Interplay of pericentromeric genome organization and chromatin landscape regulates the expression of Drosophila melanogaster heterochromatic genes. Epigenetics Chromatin 2020; 13:41. [PMID: 33028366 PMCID: PMC7541242 DOI: 10.1186/s13072-020-00358-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 09/15/2020] [Indexed: 01/04/2023] Open
Abstract
Background Transcription of genes residing within constitutive heterochromatin is paradoxical to the tenets of epigenetic code. The regulatory mechanisms of Drosophila melanogaster heterochromatic gene transcription remain largely unknown. Emerging evidence suggests that genome organization and transcriptional regulation are inter-linked. However, the pericentromeric genome organization is relatively less studied. Therefore, we sought to characterize the pericentromeric genome organization and understand how this organization along with the pericentromeric factors influences heterochromatic gene expression. Results Here, we characterized the pericentromeric genome organization in Drosophila melanogaster using 5C sequencing. Heterochromatic topologically associating domains (Het TADs) correlate with distinct epigenomic domains of active and repressed heterochromatic genes at the pericentromeres. These genes are known to depend on the heterochromatic landscape for their expression. However, HP1a or Su(var)3-9 RNAi has minimal effects on heterochromatic gene expression, despite causing significant changes in the global Het TAD organization. Probing further into this observation, we report the role of two other chromatin proteins enriched at the pericentromeres-dMES-4 and dADD1 in regulating the expression of a subset of heterochromatic genes. Conclusions Distinct pericentromeric genome organization and chromatin landscapes maintained by the interplay of heterochromatic factors (HP1a, H3K9me3, dMES-4 and dADD1) are sufficient to support heterochromatic gene expression despite the loss of global Het TAD structure. These findings open new avenues for future investigations into the mechanisms of heterochromatic gene expression.
Collapse
|
12
|
Torres DE, Oggenfuss U, Croll D, Seidl MF. Genome evolution in fungal plant pathogens: looking beyond the two-speed genome model. FUNGAL BIOL REV 2020. [DOI: 10.1016/j.fbr.2020.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
13
|
Liu J, Ali M, Zhou Q. Establishment and evolution of heterochromatin. Ann N Y Acad Sci 2020; 1476:59-77. [PMID: 32017156 PMCID: PMC7586837 DOI: 10.1111/nyas.14303] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/31/2019] [Accepted: 01/02/2020] [Indexed: 12/12/2022]
Abstract
The eukaryotic genome is packaged into transcriptionally active euchromatin and silent heterochromatin, with most studies focused on the former encompassing the majority of protein-coding genes. The recent development of various sequencing techniques has refined this classic dichromatic partition and has better illuminated the composition, establishment, and evolution of this genomic and epigenomic "dark matter" in the context of topologically associated domains and phase-separated droplets. Heterochromatin includes genomic regions that can be densely stained by chemical dyes, which have been shown to be enriched for repetitive elements and epigenetic marks, including H3K9me2/3 and H3K27me3. Heterochromatin is usually replicated late, concentrated at the nuclear periphery or around nucleoli, and usually lacks highly expressed genes; and now it is considered to be as neither genetically inert nor developmentally static. Heterochromatin guards genome integrity against transposon activities and exerts important regulatory functions by targeting beyond its contained genes. Both its nucleotide sequences and regulatory proteins exhibit rapid coevolution between species. In addition, there are dynamic transitions between euchromatin and heterochromatin during developmental and evolutionary processes. We summarize here the ever-changing characteristics of heterochromatin and propose models and principles for the evolutionary transitions of heterochromatin that have been mainly learned from studies of Drosophila and yeast. Finally, we highlight the role of sex chromosomes in studying heterochromatin evolution.
Collapse
Affiliation(s)
- Jing Liu
- MOE Laboratory of Biosystems Homeostasis & Protection, Life Sciences InstituteZhejiang UniversityHangzhouChina
- Department of Molecular Evolution and DevelopmentUniversity of ViennaViennaAustria
| | - Mujahid Ali
- Department of Molecular Evolution and DevelopmentUniversity of ViennaViennaAustria
| | - Qi Zhou
- MOE Laboratory of Biosystems Homeostasis & Protection, Life Sciences InstituteZhejiang UniversityHangzhouChina
- Department of Molecular Evolution and DevelopmentUniversity of ViennaViennaAustria
- Center for Reproductive Medicine, The 2nd Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
14
|
Funikov SY, Rezvykh AP, Kulikova DA, Zelentsova ES, Protsenko LA, Chuvakova LN, Tyukmaeva VI, Arkhipova IR, Evgen'ev MB. Adaptation of gene loci to heterochromatin in the course of Drosophila evolution is associated with insulator proteins. Sci Rep 2020; 10:11893. [PMID: 32681087 PMCID: PMC7368049 DOI: 10.1038/s41598-020-68879-2] [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: 01/10/2020] [Accepted: 06/23/2020] [Indexed: 01/11/2023] Open
Abstract
Pericentromeric heterochromatin is generally composed of repetitive DNA forming a transcriptionally repressive environment. Dozens of genes were embedded into pericentromeric heterochromatin during evolution of Drosophilidae lineage while retaining activity. However, factors that contribute to insusceptibility of gene loci to transcriptional silencing remain unknown. Here, we find that the promoter region of genes that can be embedded in both euchromatin and heterochromatin exhibits a conserved structure throughout the Drosophila phylogeny and carries motifs for binding of certain chromatin remodeling factors, including insulator proteins. Using ChIP-seq data, we demonstrate that evolutionary gene relocation between euchromatin and pericentric heterochromatin occurred with preservation of sites of insulation of BEAF-32 in evolutionarily distant species, i.e. D. melanogaster and D. virilis. Moreover, promoters of virtually all protein-coding genes located in heterochromatin in D. melanogaster are enriched with insulator proteins BEAF-32, GAF and dCTCF. Applying RNA-seq of a BEAF-32 mutant, we show that the impairment of BEAF-32 function has a complex effect on gene expression in D. melanogaster, affecting even those genes that lack BEAF-32 association in their promoters. We propose that conserved intrinsic properties of genes, such as sites of insulation near the promoter regions, may contribute to adaptation of genes to the heterochromatic environment and, hence, facilitate the evolutionary relocation of genes loci between euchromatin and heterochromatin.
Collapse
Affiliation(s)
- Sergei Yu Funikov
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, 119991, Russia
| | - Alexander P Rezvykh
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, 119991, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia
| | - Dina A Kulikova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - Elena S Zelentsova
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, 119991, Russia
| | - Lyudmila A Protsenko
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, 119991, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia
| | - Lyubov N Chuvakova
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, 119991, Russia
| | - Venera I Tyukmaeva
- Department of Biological and Environmental Science, University of Jyväskylä, 40014, Jyväskylä, Finland
| | - Irina R Arkhipova
- Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, Marine Biological Laboratory, Woods Hole, MA, USA
| | - Michael B Evgen'ev
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, 119991, Russia.
| |
Collapse
|
15
|
Moschetti R, Palazzo A, Lorusso P, Viggiano L, Massimiliano Marsano R. "What You Need, Baby, I Got It": Transposable Elements as Suppliers of Cis-Operating Sequences in Drosophila. BIOLOGY 2020; 9:E25. [PMID: 32028630 PMCID: PMC7168160 DOI: 10.3390/biology9020025] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/27/2020] [Accepted: 01/30/2020] [Indexed: 12/18/2022]
Abstract
Transposable elements (TEs) are constitutive components of both eukaryotic and prokaryotic genomes. The role of TEs in the evolution of genes and genomes has been widely assessed over the past years in a variety of model and non-model organisms. Drosophila is undoubtedly among the most powerful model organisms used for the purpose of studying the role of transposons and their effects on the stability and evolution of genes and genomes. Besides their most intuitive role as insertional mutagens, TEs can modify the transcriptional pattern of host genes by juxtaposing new cis-regulatory sequences. A key element of TE biology is that they carry transcriptional control elements that fine-tune the transcription of their own genes, but that can also perturb the transcriptional activity of neighboring host genes. From this perspective, the transposition-mediated modulation of gene expression is an important issue for the short-term adaptation of physiological functions to the environmental changes, and for long-term evolutionary changes. Here, we review the current literature concerning the regulatory and structural elements operating in cis provided by TEs in Drosophila. Furthermore, we highlight that, besides their influence on both TEs and host genes expression, they can affect the chromatin structure and epigenetic status as well as both the chromosome's structure and stability. It emerges that Drosophila is a good model organism to study the effect of TE-linked regulatory sequences, and it could help future studies on TE-host interactions in any complex eukaryotic genome.
Collapse
Affiliation(s)
- Roberta Moschetti
- Dipartimento di Biologia, Università degli Studi di Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy; (R.M.); (P.L.); (L.V.)
| | - Antonio Palazzo
- Laboratory of Translational Nanotechnology, “Istituto Tumori Giovanni Paolo II” I.R.C.C.S, Viale Orazio Flacco 65, 70125 Bari, Italy;
| | - Patrizio Lorusso
- Dipartimento di Biologia, Università degli Studi di Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy; (R.M.); (P.L.); (L.V.)
| | - Luigi Viggiano
- Dipartimento di Biologia, Università degli Studi di Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy; (R.M.); (P.L.); (L.V.)
| | - René Massimiliano Marsano
- Dipartimento di Biologia, Università degli Studi di Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy; (R.M.); (P.L.); (L.V.)
| |
Collapse
|
16
|
Saha P, Mishra RK. Heterochromatic hues of transcription-the diverse roles of noncoding transcripts from constitutive heterochromatin. FEBS J 2019; 286:4626-4641. [PMID: 31644838 DOI: 10.1111/febs.15104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/19/2019] [Accepted: 10/22/2019] [Indexed: 02/05/2023]
Abstract
Constitutive heterochromatin has been canonically considered as transcriptionally inert chromosomal regions, which silences the repeats and transposable elements (TEs), to preserve genomic integrity. However, several studies from the last few decades show that centromeric and pericentromeric regions also get transcribed and these transcripts are involved in multiple cellular processes. Regulation of such spatially and temporally controlled transcription and their relevance to heterochromatin function have emerged as an active area of research in chromatin biology. Here, we review the myriad of roles of noncoding transcripts from the constitutive heterochromatin in the establishment and maintenance of heterochromatin, kinetochore assembly, germline epigenome maintenance, early development, and diseases. Contrary to general expectations, there are active protein-coding genes in the heterochromatin although the regulatory mechanisms of their expression are largely unknown. We propose plausible hypotheses to explain heterochromatic gene expression using Drosophila melanogaster as a model, and discuss the evolutionary significance of these transcripts in the context of Drosophilid speciation. Such analyses offer insights into the regulatory pathways and functions of heterochromatic transcripts which open new avenues for further investigation.
Collapse
Affiliation(s)
- Parna Saha
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rakesh K Mishra
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
17
|
A New Portrait of Constitutive Heterochromatin: Lessons from Drosophila melanogaster. Trends Genet 2019; 35:615-631. [PMID: 31320181 DOI: 10.1016/j.tig.2019.06.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 12/14/2022]
Abstract
Constitutive heterochromatin represents a significant portion of eukaryotic genomes, but its functions still need to be elucidated. Even in the most updated genetics and molecular biology textbooks, constitutive heterochromatin is portrayed mainly as the 'silent' component of eukaryotic genomes. However, there may be more complexity to the relationship between heterochromatin and gene expression. In the fruit fly Drosophila melanogaster, a model for heterochromatin studies, about one-third of the genome is heterochromatic and is concentrated in the centric, pericentric, and telomeric regions of the chromosomes. Recent findings indicate that hundreds of D. melanogaster genes can 'live and work' properly within constitutive heterochromatin. The genomic size of these genes is generally larger than that of euchromatic genes and together they account for a significant fraction of the entire constitutive heterochromatin. Thus, this peculiar genome component in spite its ability to induce silencing, has in fact the means for being quite dynamic. A major scope of this review is to revisit the 'dogma of silent heterochromatin'.
Collapse
|
18
|
Saha P, Sowpati DT, Mishra RK. Epigenomic and genomic landscape of Drosophila melanogaster heterochromatic genes. Genomics 2019; 111:177-185. [DOI: 10.1016/j.ygeno.2018.02.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 01/21/2018] [Accepted: 02/04/2018] [Indexed: 01/05/2023]
|
19
|
Zhao PA, Rivera-Mulia JC, Gilbert DM. Replication Domains: Genome Compartmentalization into Functional Replication Units. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1042:229-257. [DOI: 10.1007/978-981-10-6955-0_11] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
20
|
Leung W, Shaffer CD, Chen EJ, Quisenberry TJ, Ko K, Braverman JM, Giarla TC, Mortimer NT, Reed LK, Smith ST, Robic S, McCartha SR, Perry DR, Prescod LM, Sheppard ZA, Saville KJ, McClish A, Morlock EA, Sochor VR, Stanton B, Veysey-White IC, Revie D, Jimenez LA, Palomino JJ, Patao MD, Patao SM, Himelblau ET, Campbell JD, Hertz AL, McEvilly MF, Wagner AR, Youngblom J, Bedi B, Bettincourt J, Duso E, Her M, Hilton W, House S, Karimi M, Kumimoto K, Lee R, Lopez D, Odisho G, Prasad R, Robbins HL, Sandhu T, Selfridge T, Tsukashima K, Yosif H, Kokan NP, Britt L, Zoellner A, Spana EP, Chlebina BT, Chong I, Friedman H, Mammo DA, Ng CL, Nikam VS, Schwartz NU, Xu TQ, Burg MG, Batten SM, Corbeill LM, Enoch E, Ensign JJ, Franks ME, Haiker B, Ingles JA, Kirkland LD, Lorenz-Guertin JM, Matthews J, Mittig CM, Monsma N, Olson KJ, Perez-Aragon G, Ramic A, Ramirez JR, Scheiber C, Schneider PA, Schultz DE, Simon M, Spencer E, Wernette AC, Wykle ME, Zavala-Arellano E, McDonald MJ, Ostby K, Wendland P, DiAngelo JR, Ceasrine AM, Cox AH, Docherty JEB, Gingras RM, Grieb SM, Pavia MJ, Personius CL, Polak GL, Beach DL, Cerritos HL, Horansky EA, Sharif KA, Moran R, Parrish S, Bickford K, Bland J, Broussard J, Campbell K, Deibel KE, Forka R, Lemke MC, Nelson MB, O'Keeffe C, Ramey SM, Schmidt L, Villegas P, Jones CJ, Christ SL, Mamari S, Rinaldi AS, Stity G, Hark AT, Scheuerman M, Silver Key SC, McRae BD, Haberman AS, Asinof S, Carrington H, Drumm K, Embry T, McGuire R, Miller-Foreman D, Rosen S, Safa N, Schultz D, Segal M, Shevin Y, Svoronos P, Vuong T, Skuse G, Paetkau DW, Bridgman RK, Brown CM, Carroll AR, Gifford FM, Gillespie JB, Herman SE, Holtcamp KL, Host MA, Hussey G, Kramer DM, Lawrence JQ, Martin MM, Niemiec EN, O'Reilly AP, Pahl OA, Quintana G, Rettie EAS, Richardson TL, Rodriguez AE, Rodriguez MO, Schiraldi L, Smith JJ, Sugrue KF, Suriano LJ, Takach KE, Vasquez AM, Velez X, Villafuerte EJ, Vives LT, Zellmer VR, Hauke J, Hauser CR, Barker K, Cannon L, Parsamian P, Parsons S, Wichman Z, Bazinet CW, Johnson DE, Bangura A, Black JA, Chevee V, Einsteen SA, Hilton SK, Kollmer M, Nadendla R, Stamm J, Fafara-Thompson AE, Gygi AM, Ogawa EE, Van Camp M, Kocsisova Z, Leatherman JL, Modahl CM, Rubin MR, Apiz-Saab SS, Arias-Mejias SM, Carrion-Ortiz CF, Claudio-Vazquez PN, Espada-Green DM, Feliciano-Camacho M, Gonzalez-Bonilla KM, Taboas-Arroyo M, Vargas-Franco D, Montañez-Gonzalez R, Perez-Otero J, Rivera-Burgos M, Rivera-Rosario FJ, Eisler HL, Alexander J, Begley SK, Gabbard D, Allen RJ, Aung WY, Barshop WD, Boozalis A, Chu VP, Davis JS, Duggal RN, Franklin R, Gavinski K, Gebreyesus H, Gong HZ, Greenstein RA, Guo AD, Hanson C, Homa KE, Hsu SC, Huang Y, Huo L, Jacobs S, Jia S, Jung KL, Wai-Chee Kong S, Kroll MR, Lee BM, Lee PF, Levine KM, Li AS, Liu C, Liu MM, Lousararian AP, Lowery PB, Mallya AP, Marcus JE, Ng PC, Nguyen HP, Patel R, Precht H, Rastogi S, Sarezky JM, Schefkind A, Schultz MB, Shen D, Skorupa T, Spies NC, Stancu G, Vivian Tsang HM, Turski AL, Venkat R, Waldman LE, Wang K, Wang T, Wei JW, Wu DY, Xiong DD, Yu J, Zhou K, McNeil GP, Fernandez RW, Menzies PG, Gu T, Buhler J, Mardis ER, Elgin SCR. Retrotransposons Are the Major Contributors to the Expansion of the Drosophila ananassae Muller F Element. G3 (BETHESDA, MD.) 2017; 7:2439-2460. [PMID: 28667019 PMCID: PMC5555453 DOI: 10.1534/g3.117.040907] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 04/03/2017] [Indexed: 11/24/2022]
Abstract
The discordance between genome size and the complexity of eukaryotes can partly be attributed to differences in repeat density. The Muller F element (∼5.2 Mb) is the smallest chromosome in Drosophila melanogaster, but it is substantially larger (>18.7 Mb) in D. ananassae To identify the major contributors to the expansion of the F element and to assess their impact, we improved the genome sequence and annotated the genes in a 1.4-Mb region of the D. ananassae F element, and a 1.7-Mb region from the D element for comparison. We find that transposons (particularly LTR and LINE retrotransposons) are major contributors to this expansion (78.6%), while Wolbachia sequences integrated into the D. ananassae genome are minor contributors (0.02%). Both D. melanogaster and D. ananassae F-element genes exhibit distinct characteristics compared to D-element genes (e.g., larger coding spans, larger introns, more coding exons, and lower codon bias), but these differences are exaggerated in D. ananassae Compared to D. melanogaster, the codon bias observed in D. ananassae F-element genes can primarily be attributed to mutational biases instead of selection. The 5' ends of F-element genes in both species are enriched in dimethylation of lysine 4 on histone 3 (H3K4me2), while the coding spans are enriched in H3K9me2. Despite differences in repeat density and gene characteristics, D. ananassae F-element genes show a similar range of expression levels compared to genes in euchromatic domains. This study improves our understanding of how transposons can affect genome size and how genes can function within highly repetitive domains.
Collapse
Affiliation(s)
- Wilson Leung
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | | | - Elizabeth J Chen
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | | | - Kevin Ko
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - John M Braverman
- Department of Biology, Saint Joseph's University, Philadelphia, PA 19131
| | | | - Nathan T Mortimer
- School of Biological Sciences, Illinois State University, Normal, IL 61790
| | - Laura K Reed
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35401
| | - Sheryl T Smith
- Department of Biology, Arcadia University, Glenside, PA 19038
| | - Srebrenka Robic
- Department of Biology, Agnes Scott College, Decatur, GA 30030
| | | | | | | | | | - Ken J Saville
- Department of Biology, Albion College, Albion, MI 49224
| | | | | | | | | | | | - Dennis Revie
- Department of Biology, California Lutheran University, Thousand Oaks, CA 91360
| | - Luis A Jimenez
- Department of Biology, California Lutheran University, Thousand Oaks, CA 91360
| | - Jennifer J Palomino
- Department of Biology, California Lutheran University, Thousand Oaks, CA 91360
| | - Melissa D Patao
- Department of Biology, California Lutheran University, Thousand Oaks, CA 91360
| | - Shane M Patao
- Department of Biology, California Lutheran University, Thousand Oaks, CA 91360
| | - Edward T Himelblau
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93405
| | - Jaclyn D Campbell
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93405
| | - Alexandra L Hertz
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93405
| | - Maddison F McEvilly
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93405
| | - Allison R Wagner
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93405
| | - James Youngblom
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Baljit Bedi
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Jeffery Bettincourt
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Erin Duso
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Maiye Her
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - William Hilton
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Samantha House
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Masud Karimi
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Kevin Kumimoto
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Rebekah Lee
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Darryl Lopez
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - George Odisho
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Ricky Prasad
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Holly Lyn Robbins
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Tanveer Sandhu
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Tracy Selfridge
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Kara Tsukashima
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Hani Yosif
- Department of Biology, California State University, Stanislaus, Turlock, CA 95382
| | - Nighat P Kokan
- Department of Natural Sciences, Cardinal Stritch University, Milwaukee, WI 53217
| | - Latia Britt
- Department of Natural Sciences, Cardinal Stritch University, Milwaukee, WI 53217
| | - Alycia Zoellner
- Department of Natural Sciences, Cardinal Stritch University, Milwaukee, WI 53217
| | - Eric P Spana
- Department of Biology, Duke University, Durham, NC 27708
| | - Ben T Chlebina
- Department of Biology, Duke University, Durham, NC 27708
| | - Insun Chong
- Department of Biology, Duke University, Durham, NC 27708
| | | | - Danny A Mammo
- Department of Biology, Duke University, Durham, NC 27708
| | - Chun L Ng
- Department of Biology, Duke University, Durham, NC 27708
| | | | | | - Thomas Q Xu
- Department of Biology, Duke University, Durham, NC 27708
| | - Martin G Burg
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Spencer M Batten
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Lindsay M Corbeill
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Erica Enoch
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Jesse J Ensign
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Mary E Franks
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Breanna Haiker
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Judith A Ingles
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Lyndsay D Kirkland
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Joshua M Lorenz-Guertin
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Jordan Matthews
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Cody M Mittig
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Nicholaus Monsma
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Katherine J Olson
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Guillermo Perez-Aragon
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Alen Ramic
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Jordan R Ramirez
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Christopher Scheiber
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Patrick A Schneider
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Devon E Schultz
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Matthew Simon
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Eric Spencer
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Adam C Wernette
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Maxine E Wykle
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Elizabeth Zavala-Arellano
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Mitchell J McDonald
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Kristine Ostby
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | - Peter Wendland
- Departments of Biomedical Sciences and Cell and Molecular Biology, Grand Valley State University, Allendale, MI 49401
| | | | | | - Amanda H Cox
- Department of Biology, Hofstra University, Hempstead, NY 11549
| | | | | | | | - Michael J Pavia
- Department of Biology, Hofstra University, Hempstead, NY 11549
| | | | | | - Dale L Beach
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA 23909
| | - Heaven L Cerritos
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA 23909
| | - Edward A Horansky
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA 23909
| | - Karim A Sharif
- Department of Biology, Massasoit Community College, Brockton, MA 02302
| | - Ryan Moran
- Department of Biology, Massasoit Community College, Brockton, MA 02302
| | - Susan Parrish
- Department of Biology, McDaniel College, Westminster, MD 21157
| | | | - Jennifer Bland
- Department of Biology, McDaniel College, Westminster, MD 21157
| | | | - Kerry Campbell
- Department of Biology, McDaniel College, Westminster, MD 21157
| | | | - Richard Forka
- Department of Biology, McDaniel College, Westminster, MD 21157
| | - Monika C Lemke
- Department of Biology, McDaniel College, Westminster, MD 21157
| | - Marlee B Nelson
- Department of Biology, McDaniel College, Westminster, MD 21157
| | | | - S Mariel Ramey
- Department of Biology, McDaniel College, Westminster, MD 21157
| | - Luke Schmidt
- Department of Biology, McDaniel College, Westminster, MD 21157
| | - Paola Villegas
- Department of Biology, McDaniel College, Westminster, MD 21157
| | | | - Stephanie L Christ
- Department of Biological Sciences, Moravian College, Bethlehem, PA 18018
| | - Sami Mamari
- Department of Biological Sciences, Moravian College, Bethlehem, PA 18018
| | - Adam S Rinaldi
- Department of Biological Sciences, Moravian College, Bethlehem, PA 18018
| | - Ghazal Stity
- Department of Biological Sciences, Moravian College, Bethlehem, PA 18018
| | - Amy T Hark
- Department of Biology, Muhlenberg College, Allentown, PA 18104
| | - Mark Scheuerman
- Department of Biology, Muhlenberg College, Allentown, PA 18104
| | - S Catherine Silver Key
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707
| | - Briana D McRae
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707
| | | | - Sam Asinof
- Department of Biology, Oberlin College, Oberlin, OH 44074
| | | | - Kelly Drumm
- Department of Biology, Oberlin College, Oberlin, OH 44074
| | - Terrance Embry
- Department of Biology, Oberlin College, Oberlin, OH 44074
| | | | | | - Stella Rosen
- Department of Biology, Oberlin College, Oberlin, OH 44074
| | - Nadia Safa
- Department of Biology, Oberlin College, Oberlin, OH 44074
| | - Darrin Schultz
- Department of Biology, Oberlin College, Oberlin, OH 44074
| | - Matt Segal
- Department of Biology, Oberlin College, Oberlin, OH 44074
| | - Yakov Shevin
- Department of Biology, Oberlin College, Oberlin, OH 44074
| | | | - Tam Vuong
- Department of Biology, Oberlin College, Oberlin, OH 44074
| | - Gary Skuse
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY 14623
| | - Don W Paetkau
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | | | | | - Alicia R Carroll
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | | | | | - Susan E Herman
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | | | - Misha A Host
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | - Gabrielle Hussey
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | | | - Joan Q Lawrence
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | | | - Ellen N Niemiec
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | | | - Olivia A Pahl
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | | | | | | | | | - Mona O Rodriguez
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | - Laura Schiraldi
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | - Joanna J Smith
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | - Kelsey F Sugrue
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | | | - Kaitlyn E Takach
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | | | - Ximena Velez
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | | | - Laura T Vives
- Department of Biology, Saint Mary's College, Notre Dame, IN 46556
| | | | - Jeanette Hauke
- Department of Biology, Simmons College, Boston, MA 02115
| | - Charles R Hauser
- Bioinformatics Program, St. Edward's University, Austin, TX 78704
| | - Karolyn Barker
- Bioinformatics Program, St. Edward's University, Austin, TX 78704
| | - Laurie Cannon
- Bioinformatics Program, St. Edward's University, Austin, TX 78704
| | | | - Samantha Parsons
- Bioinformatics Program, St. Edward's University, Austin, TX 78704
| | | | | | - Diana E Johnson
- Department of Biological Sciences, The George Washington University, Washington, DC 20052
| | - Abubakarr Bangura
- Department of Biological Sciences, The George Washington University, Washington, DC 20052
| | - Jordan A Black
- Department of Biological Sciences, The George Washington University, Washington, DC 20052
| | - Victoria Chevee
- Department of Biological Sciences, The George Washington University, Washington, DC 20052
| | - Sarah A Einsteen
- Department of Biological Sciences, The George Washington University, Washington, DC 20052
| | - Sarah K Hilton
- Department of Biological Sciences, The George Washington University, Washington, DC 20052
| | - Max Kollmer
- Department of Biological Sciences, The George Washington University, Washington, DC 20052
| | - Rahul Nadendla
- Department of Biological Sciences, The George Washington University, Washington, DC 20052
| | - Joyce Stamm
- Department of Biology, University of Evansville, Evansville, IN 47722
| | | | - Amber M Gygi
- Department of Biology, University of Evansville, Evansville, IN 47722
| | - Emmy E Ogawa
- Department of Biology, University of Evansville, Evansville, IN 47722
| | - Matt Van Camp
- Department of Biology, University of Evansville, Evansville, IN 47722
| | - Zuzana Kocsisova
- Department of Biology, University of Evansville, Evansville, IN 47722
| | - Judith L Leatherman
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639
| | - Cassie M Modahl
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639
| | - Michael R Rubin
- Department of Biology, University of Puerto Rico at Cayey, Cayey, PR 00736
| | - Susana S Apiz-Saab
- Department of Biology, University of Puerto Rico at Cayey, Cayey, PR 00736
| | | | | | | | | | | | | | | | | | | | - Joseph Perez-Otero
- Department of Biology, University of Puerto Rico at Cayey, Cayey, PR 00736
| | | | | | - Heather L Eisler
- Department of Biology, University of the Cumberlands, Williamsburg, KY 40769
| | - Jackie Alexander
- Department of Biology, University of the Cumberlands, Williamsburg, KY 40769
| | - Samatha K Begley
- Department of Biology, University of the Cumberlands, Williamsburg, KY 40769
| | - Deana Gabbard
- Department of Biology, University of the Cumberlands, Williamsburg, KY 40769
| | - Robert J Allen
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Wint Yan Aung
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - William D Barshop
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Amanda Boozalis
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Vanessa P Chu
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Jeremy S Davis
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Ryan N Duggal
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Robert Franklin
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Katherine Gavinski
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Heran Gebreyesus
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Henry Z Gong
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Rachel A Greenstein
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Averill D Guo
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Casey Hanson
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Kaitlin E Homa
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Simon C Hsu
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Yi Huang
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Lucy Huo
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Sarah Jacobs
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Sasha Jia
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Kyle L Jung
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Sarah Wai-Chee Kong
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Matthew R Kroll
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Brandon M Lee
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Paul F Lee
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Kevin M Levine
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Amy S Li
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Chengyu Liu
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Max Mian Liu
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Adam P Lousararian
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Peter B Lowery
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Allyson P Mallya
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Joseph E Marcus
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Patrick C Ng
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Hien P Nguyen
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Ruchik Patel
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Hashini Precht
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Suchita Rastogi
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Jonathan M Sarezky
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Adam Schefkind
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Michael B Schultz
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Delia Shen
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Tara Skorupa
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Nicholas C Spies
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Gabriel Stancu
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | | | - Alice L Turski
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Rohit Venkat
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Leah E Waldman
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Kaidi Wang
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Tracy Wang
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Jeffrey W Wei
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Dennis Y Wu
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - David D Xiong
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Jack Yu
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Karen Zhou
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Gerard P McNeil
- Department of Biology, York College / CUNY, Jamaica, NY 11451
| | | | | | - Tingting Gu
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| | - Jeremy Buhler
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130
| | - Elaine R Mardis
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108
| | - Sarah C R Elgin
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130
| |
Collapse
|
21
|
Caizzi R, Moschetti R, Piacentini L, Fanti L, Marsano RM, Dimitri P. Comparative Genomic Analyses Provide New Insights into the Evolutionary Dynamics of Heterochromatin in Drosophila. PLoS Genet 2016; 12:e1006212. [PMID: 27513559 PMCID: PMC4981424 DOI: 10.1371/journal.pgen.1006212] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 07/02/2016] [Indexed: 12/21/2022] Open
Abstract
The term heterochromatin has been long considered synonymous with gene silencing, but it is now clear that the presence of transcribed genes embedded in pericentromeric heterochromatin is a conserved feature in the evolution of eukaryotic genomes. Several studies have addressed the epigenetic changes that enable the expression of genes in pericentric heterochromatin, yet little is known about the evolutionary processes through which this has occurred. By combining genome annotation analysis and high-resolution cytology, we have identified and mapped 53 orthologs of D. melanogaster heterochromatic genes in the genomes of two evolutionarily distant species, D. pseudoobscura and D. virilis. Our results show that the orthologs of the D. melanogaster heterochromatic genes are clustered at three main genomic regions in D. virilis and D. pseudoobscura. In D. virilis, the clusters lie in the middle of euchromatin, while those in D. pseudoobscura are located in the proximal portion of the chromosome arms. Some orthologs map to the corresponding Muller C element in D. pseudoobscura and D. virilis, while others localize on the Muller B element, suggesting that chromosomal rearrangements that have been instrumental in the fusion of two separate elements involved the progenitors of genes currently located in D. melanogaster heterochromatin. These results demonstrate an evolutionary repositioning of gene clusters from ancestral locations in euchromatin to the pericentromeric heterochromatin of descendent D. melanogaster chromosomes. Remarkably, in both D. virilis and D. pseudoobscura the gene clusters show a conserved association with the HP1a protein, one of the most highly evolutionarily conserved epigenetic marks. In light of these results, we suggest a new scenario whereby ancestral HP1-like proteins (and possibly other epigenetic marks) may have contributed to the evolutionary repositioning of gene clusters into heterochromatin.
Collapse
Affiliation(s)
- Ruggiero Caizzi
- Dipartimento di Biologia, Università degli Studi di Bari, Bari, Italy
- * E-mail: (RC); (PD)
| | - Roberta Moschetti
- Dipartimento di Biologia, Università degli Studi di Bari, Bari, Italy
| | - Lucia Piacentini
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie ‘‘Charles Darwin”, Sapienza Università di Roma, Roma, Italy
| | - Laura Fanti
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie ‘‘Charles Darwin”, Sapienza Università di Roma, Roma, Italy
| | | | - Patrizio Dimitri
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie ‘‘Charles Darwin”, Sapienza Università di Roma, Roma, Italy
- * E-mail: (RC); (PD)
| |
Collapse
|
22
|
Moschetti R, Celauro E, Cruciani F, Caizzi R, Dimitri P. On the evolution of Yeti, a Drosophila melanogaster heterochromatin gene. PLoS One 2014; 9:e113010. [PMID: 25405891 PMCID: PMC4236135 DOI: 10.1371/journal.pone.0113010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 10/22/2014] [Indexed: 11/22/2022] Open
Abstract
Constitutive heterochromatin is a ubiquitous and still unveiled component of eukaryotic genomes, within which it comprises large portions. Although constitutive heterochromatin is generally considered to be transcriptionally silent, it contains a significant variety of sequences that are expressed, among which about 300 single-copy coding genes have been identified by genetic and genomic analyses in the last decades. Here, we report the results of the evolutionary analysis of Yeti, an essential gene of Drosophila melanogaster located in the deep pericentromeric region of chromosome 2R. By FISH, we showed that Yeti maintains a heterochromatin location in both D. simulans and D. sechellia species, closely related to D. melanogaster, while in the more distant species e.g., D. pseudoobscura and D. virilis, it is found within euchromatin, in the syntenic chromosome Muller C, that corresponds to the 2R arm of D. melanogaster chromosome 2. Thus, over evolutionary time, Yeti has been resident on the same chromosomal element, but it progressively moved closer to the pericentric regions. Moreover, in silico reconstruction of the Yeti gene structure in 19 Drosophila species and in 5 non-drosophilid dipterans shows a rather stable organization during evolution. Accordingly, by PCR analysis and sequencing, we found that the single intron of Yeti does not undergo major intraspecies or interspecies size changes, unlike the introns of other essential Drosophila heterochromatin genes, such as light and Dbp80. This implicates diverse evolutionary forces in shaping the structural organization of genes found within heterochromatin. Finally, the results of dS - dN tests show that Yeti is under negative selection both in heterochromatin and euchromatin, and indicate that the change in genomic location did not affected significantly the molecular evolution of the gene. Together, the results of this work contribute to our understanding of the evolutionary dynamics of constitutive heterochromatin in the genomes of higher eukaryotes.
Collapse
Affiliation(s)
- Roberta Moschetti
- Dipartimento di Biologia, Università degli Studi di Bari, Bari, Italy
| | - Emanuele Celauro
- Dipartimento di Biologia e Biotecnologie “Charles Darwin” and Istituto Pasteur Fondazione Cenci-Bolognetti, Sapienza Università di Roma, Roma, Italy
| | - Fulvio Cruciani
- Dipartimento di Biologia e Biotecnologie “Charles Darwin” and Istituto Pasteur Fondazione Cenci-Bolognetti, Sapienza Università di Roma, Roma, Italy
| | - Ruggiero Caizzi
- Dipartimento di Biologia, Università degli Studi di Bari, Bari, Italy
| | - Patrizio Dimitri
- Dipartimento di Biologia e Biotecnologie “Charles Darwin” and Istituto Pasteur Fondazione Cenci-Bolognetti, Sapienza Università di Roma, Roma, Italy
- * E-mail:
| |
Collapse
|
23
|
Cryderman DE, Vitalini MW, Wallrath LL. Heterochromatin protein 1a is required for an open chromatin structure. Transcription 2014; 2:95-99. [PMID: 21468237 DOI: 10.4161/trns.2.2.14687] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 12/30/2010] [Accepted: 12/31/2010] [Indexed: 01/03/2023] Open
Abstract
The Drosophila melanogaster fourth chromosome contains interspersed domains of active and repressive chromatin. We investigated a stock harboring a silenced transgene inserted into Dyrk3 and near Caps-two expressed genes on chromosome four. In an HP1a-deficient background, transgene expression was activated while, paradoxically, expression of Dyrk3 and Caps was reduced. We found that the promoters of Dyrk3 and Caps contained DNase I hypersensitive sites but also possessed methylated histone H3 and HP1a, marks of repressive chromatin. In HP1a-deficient flies, the Dyrk3 and Caps promoters displayed diminished accessibility to nuclease digestion, revealing a surprising role for HP1a in opening chromatin.
Collapse
|
24
|
Mteirek R, Gueguen N, Jensen S, Brasset E, Vaury C. Drosophila heterochromatin: structure and function. CURRENT OPINION IN INSECT SCIENCE 2014; 1:19-24. [PMID: 32846725 DOI: 10.1016/j.cois.2014.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 04/23/2014] [Accepted: 04/25/2014] [Indexed: 06/11/2023]
Abstract
Heterochromatic domains, which are enriched in repetitive sequences and packaged in a higher-order chromatin folding, carry the potential to epigenetically inactivate a euchromatic gene that has been moved in close proximity. The discovery that these domains encode non-coding RNAs involved in RNA-silencing mechanisms has recently contributed to a better understanding of the mechanisms of the epigenetic repression established by heterochromatic domains. In this review, we will consider the repeated nature of their DNA sequence, the successive steps in heterochromatin assembly, starting with the decision process, the higher order state assembly and its epigenetic propagation. Recent findings provide new insights into the cellular functions of heterochromatin, notably its major contribution to genome stability and chromosome integrity.
Collapse
Affiliation(s)
- Rana Mteirek
- Clermont Université, Université d'Auvergne, Laboratoire GReD, BP 38, 63001 Clermont-Ferrand, France; Inserm, U 1103, BP 38, 63001 Clermont-Ferrand, France; CNRS, UMR 6293, BP 38, 63001 Clermont-Ferrand, France
| | - Nathalie Gueguen
- Clermont Université, Université d'Auvergne, Laboratoire GReD, BP 38, 63001 Clermont-Ferrand, France; Inserm, U 1103, BP 38, 63001 Clermont-Ferrand, France; CNRS, UMR 6293, BP 38, 63001 Clermont-Ferrand, France
| | - Silke Jensen
- Clermont Université, Université d'Auvergne, Laboratoire GReD, BP 38, 63001 Clermont-Ferrand, France; Inserm, U 1103, BP 38, 63001 Clermont-Ferrand, France; CNRS, UMR 6293, BP 38, 63001 Clermont-Ferrand, France
| | - Emilie Brasset
- Clermont Université, Université d'Auvergne, Laboratoire GReD, BP 38, 63001 Clermont-Ferrand, France; Inserm, U 1103, BP 38, 63001 Clermont-Ferrand, France; CNRS, UMR 6293, BP 38, 63001 Clermont-Ferrand, France
| | - Chantal Vaury
- Clermont Université, Université d'Auvergne, Laboratoire GReD, BP 38, 63001 Clermont-Ferrand, France; Inserm, U 1103, BP 38, 63001 Clermont-Ferrand, France; CNRS, UMR 6293, BP 38, 63001 Clermont-Ferrand, France.
| |
Collapse
|
25
|
Whole-genome sequencing of Oryza brachyantha reveals mechanisms underlying Oryza genome evolution. Nat Commun 2013; 4:1595. [PMID: 23481403 PMCID: PMC3615480 DOI: 10.1038/ncomms2596] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 02/11/2013] [Indexed: 01/12/2023] Open
Abstract
The wild species of the genus Oryza contain a largely untapped reservoir of agronomically important genes for rice improvement. Here we report the 261-Mb de novo assembled genome sequence of Oryza brachyantha. Low activity of long-terminal repeat retrotransposons and massive internal deletions of ancient long-terminal repeat elements lead to the compact genome of Oryza brachyantha. We model 32,038 protein-coding genes in the Oryza brachyantha genome, of which only 70% are located in collinear positions in comparison with the rice genome. Analysing breakpoints of non-collinear genes suggests that double-strand break repair through non-homologous end joining has an important role in gene movement and erosion of collinearity in the Oryza genomes. Transition of euchromatin to heterochromatin in the rice genome is accompanied by segmental and tandem duplications, further expanded by transposable element insertions. The high-quality reference genome sequence of Oryza brachyantha provides an important resource for functional and evolutionary studies in the genus Oryza. The wild rice species can be used as germplasm resources for this crop’s genetic improvement. Here Chen and colleagues report the de novo sequencing of the O. brachyantha genome, and identify the origin of genome size variation, the role of gene movement and its implications on heterochromatin evolution in the rice genome.
Collapse
|
26
|
Sharakhova MV, Peery A, Antonio-Nkondjio C, Xia A, Ndo C, Awono-Ambene P, Simard F, Sharakhov IV. Cytogenetic analysis of Anopheles ovengensis revealed high structural divergence of chromosomes in the Anopheles nili group. INFECTION GENETICS AND EVOLUTION 2013; 16:341-8. [PMID: 23523820 DOI: 10.1016/j.meegid.2013.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 02/25/2013] [Accepted: 03/05/2013] [Indexed: 01/15/2023]
Abstract
Cytogenetic analysis is an informative classical approach to understanding the relationships among members in a group of closely related species of mosquitoes. Anopheles ovengensis is a recently discovered species of the Anopheles nili group and is one of the important malaria vectors in the African equatorial forest. This study characterized polytene chromosomes of An. ovengensis and compared them with polytene chromosomes of An. nili. Using fluorescent in situ hybridization and chromosome banding pattern comparison we have established correspondence between chromosomal arms of An. ovengensis and An. nili. Analysis of chromosome morphology in the two species revealed a limited similarity in the banding patterns. The most extensive reorganization occurs in pericentromeric and intercalary heterochromatin. Chromosomes of An. ovengensis are joined together by a diffuse chromocenter and they have two large regions of intercalary heterochromatin in arms 2L and 3R. In contrast, the chromocenter and intercalary heterochromatin are not seen in An. nili chromosomes. Comparative analysis of the arm association suggests the occurrence of a whole-arm translocation between the two members of the group. The observed, substantial reorganizations of chromosome structure implies either a rapid rate of chromosome evolution in the An. nili group, or that the two species belong to different taxonomic groups within subgenus Cellia.
Collapse
|
27
|
Lang M, Murat S, Clark AG, Gouppil G, Blais C, Matzkin LM, Guittard É, Yoshiyama−Yanagawa T, Kataoka H, Niwa R, Lafont R, Dauphin−Villemant C, Orgogozo V. Mutations in the neverland gene turned Drosophila pachea into an obligate specialist species. Science 2012; 337:1658-61. [PMID: 23019649 PMCID: PMC4729188 DOI: 10.1126/science.1224829] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Most living species exploit a limited range of resources. However, little is known about how tight associations build up during evolution between such specialist species and the hosts they use. We examined the dependence of Drosophila pachea on its single host, the senita cactus. Several amino acid changes in the Neverland oxygenase rendered D. pachea unable to transform cholesterol into 7-dehydrocholesterol (the first reaction in the steroid hormone biosynthetic pathway in insects) and thus made D. pachea dependent on the uncommon sterols of its host plant. The neverland mutations increase survival on the cactus's unusual sterols and are in a genomic region that faced recent positive selection. This study illustrates how relatively few genetic changes in a single gene may restrict the ecological niche of a species.
Collapse
Affiliation(s)
- Michael Lang
- CNRS UMR7592, Univ Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, 15 rue Hélène Brion, 75205 Paris cedex 13, France
| | - Sophie Murat
- CNRS UMR7592, Univ Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, 15 rue Hélène Brion, 75205 Paris cedex 13, France
- UPMC, Univ Paris 06, CNRS, Bâtiment A, 7 quai Saint Bernard, 75005 Paris, France
| | - Andrew G. Clark
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY USA
| | - Géraldine Gouppil
- CNRS UMR7592, Univ Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, 15 rue Hélène Brion, 75205 Paris cedex 13, France
| | - Catherine Blais
- UPMC, Univ Paris 06, CNRS, Bâtiment A, 7 quai Saint Bernard, 75005 Paris, France
| | - Luciano M. Matzkin
- Department of Biological Sciences, University of Alabama in Huntsville, 301 Sparkman Drive, Huntsville AL 35899, USA
| | - Émilie Guittard
- UPMC, Univ Paris 06, CNRS, Bâtiment A, 7 quai Saint Bernard, 75005 Paris, France
| | - Takuji Yoshiyama−Yanagawa
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8572, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Hiroshi Kataoka
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Ryusuke Niwa
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8572, Japan
| | - René Lafont
- UPMC, Univ Paris 06, CNRS, Bâtiment A, 7 quai Saint Bernard, 75005 Paris, France
| | | | - Virginie Orgogozo
- CNRS UMR7592, Univ Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, 15 rue Hélène Brion, 75205 Paris cedex 13, France
- UPMC, Univ Paris 06, CNRS, Bâtiment A, 7 quai Saint Bernard, 75005 Paris, France
| |
Collapse
|
28
|
Shilatifard A. The COMPASS family of histone H3K4 methylases: mechanisms of regulation in development and disease pathogenesis. Annu Rev Biochem 2012; 81:65-95. [PMID: 22663077 PMCID: PMC4010150 DOI: 10.1146/annurev-biochem-051710-134100] [Citation(s) in RCA: 794] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The Saccharomyces cerevisiae Set1/COMPASS was the first histone H3 lysine 4 (H3K4) methylase identified over 10 years ago. Since then, it has been demonstrated that Set1/COMPASS and its enzymatic product, H3K4 methylation, is highly conserved across the evolutionary tree. Although there is only one COMPASS in yeast, Drosophila possesses three and humans bear six COMPASS family members, each capable of methylating H3K4 with nonredundant functions. In yeast, the histone H2B monoubiquitinase Rad6/Bre1 is required for proper H3K4 and H3K79 trimethylations. The machineries involved in this process are also highly conserved from yeast to human. In this review, the process of histone H2B monoubiquitination-dependent and -independent histone H3K4 methylation as a mark of active transcription, enhancer signatures, and developmentally poised genes is discussed. The misregulation of histone H2B monoubiquitination and H3K4 methylation result in the pathogenesis of human diseases, including cancer. Recent findings in this regard are also examined.
Collapse
Affiliation(s)
- Ali Shilatifard
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA.
| |
Collapse
|
29
|
Abstract
The genome of Drosophila is protected from DNA damage during oogenesis by a mechanism involving short RNAs. Surprisingly transcription of these RNAs requires that their DNA is associated with a histone modification usually associated with gene silencing.
Collapse
Affiliation(s)
- David J Finnegan
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, King's Buildings, Mayfield Road, Edinburgh EH9 3JR, UK.
| |
Collapse
|
30
|
Hoskins RA, Landolin JM, Brown JB, Sandler JE, Takahashi H, Lassmann T, Yu C, Booth BW, Zhang D, Wan KH, Yang L, Boley N, Andrews J, Kaufman TC, Graveley BR, Bickel PJ, Carninci P, Carlson JW, Celniker SE. Genome-wide analysis of promoter architecture in Drosophila melanogaster. Genome Res 2010; 21:182-92. [PMID: 21177961 DOI: 10.1101/gr.112466.110] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Core promoters are critical regions for gene regulation in higher eukaryotes. However, the boundaries of promoter regions, the relative rates of initiation at the transcription start sites (TSSs) distributed within them, and the functional significance of promoter architecture remain poorly understood. We produced a high-resolution map of promoters active in the Drosophila melanogaster embryo by integrating data from three independent and complementary methods: 21 million cap analysis of gene expression (CAGE) tags, 1.2 million RNA ligase mediated rapid amplification of cDNA ends (RLM-RACE) reads, and 50,000 cap-trapped expressed sequence tags (ESTs). We defined 12,454 promoters of 8037 genes. Our analysis indicates that, due to non-promoter-associated RNA background signal, previous studies have likely overestimated the number of promoter-associated CAGE clusters by fivefold. We show that TSS distributions form a complex continuum of shapes, and that promoters active in the embryo and adult have highly similar shapes in 95% of cases. This suggests that these distributions are generally determined by static elements such as local DNA sequence and are not modulated by dynamic signals such as histone modifications. Transcription factor binding motifs are differentially enriched as a function of promoter shape, and peaked promoter shape is correlated with both temporal and spatial regulation of gene expression. Our results contribute to the emerging view that core promoters are functionally diverse and control patterning of gene expression in Drosophila and mammals.
Collapse
Affiliation(s)
- Roger A Hoskins
- Department of Genome Dynamics, Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 97420, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Raghavan K, Ruskin HJ, Perrin D, Goasmat F, Burns J. Computational micromodel for epigenetic mechanisms. PLoS One 2010; 5:e14031. [PMID: 21152421 PMCID: PMC2994705 DOI: 10.1371/journal.pone.0014031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 10/26/2010] [Indexed: 11/19/2022] Open
Abstract
Characterization of the epigenetic profile of humans since the initial breakthrough on the human genome project has strongly established the key role of histone modifications and DNA methylation. These dynamic elements interact to determine the normal level of expression or methylation status of the constituent genes in the genome. Recently, considerable evidence has been put forward to demonstrate that environmental stress implicitly alters epigenetic patterns causing imbalance that can lead to cancer initiation. This chain of consequences has motivated attempts to computationally model the influence of histone modification and DNA methylation in gene expression and investigate their intrinsic interdependency. In this paper, we explore the relation between DNA methylation and transcription and characterize in detail the histone modifications for specific DNA methylation levels using a stochastic approach.
Collapse
Affiliation(s)
- Karthika Raghavan
- Centre for Scientific Computing and Complex Systems Modeling, Dublin City University, Dublin, Ireland.
| | | | | | | | | |
Collapse
|
32
|
Rybina OY, Pasyukova EG. A naturally occurring polymorphism at Drosophila melanogaster Lim3 Locus, a homolog of human LHX3/4, affects Lim3 transcription and fly lifespan. PLoS One 2010; 5:e12621. [PMID: 20838645 PMCID: PMC2935391 DOI: 10.1371/journal.pone.0012621] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2010] [Accepted: 08/05/2010] [Indexed: 11/18/2022] Open
Abstract
Lim3 encodes an RNA polymerase II transcription factor with a key role in neuron specification. It was also identified as a candidate gene that affects lifespan. These pleiotropic effects indicate the fundamental significance of the potential interplay between neural development and lifespan control. The goal of this study was to analyze the causal relationships between Lim3 structural variations, and gene expression and lifespan changes, and to provide insights into regulatory pathways controlling lifespan. Fifty substitution lines containing second chromosomes from a Drosophila natural population were used to analyze the association between lifespan and sequence variation in the 5'-regulatory region, and first exon and intron of Lim3A, in which we discovered multiple transcription start sites (TSS). The core and proximal promoter organization for Lim3A and a previously unknown mRNA named Lim3C were described. A haplotype of two markers in the Lim3A regulatory region was significantly associated with variation in lifespan. We propose that polymorphisms in the regulatory region affect gene transcription, and consequently lifespan. Indeed, five polymorphic markers located within 380 to 680 bp of the Lim3A major TSS, including two markers associated with lifespan variation, were significantly associated with the level of Lim3A transcript, as evaluated by real time RT-PCR in embryos, adult heads, and testes. A naturally occurring polymorphism caused a six-fold change in gene transcription and a 25% change in lifespan. Markers associated with long lifespan and intermediate Lim3A transcription were present in the population at high frequencies. We hypothesize that polymorphic markers associated with Lim3A expression are located within the binding sites for proteins that regulate gene function, and provide general rather than tissue-specific regulation of transcription, and that intermediate levels of Lim3A expression confer a selective advantage and longer lifespan.
Collapse
|
33
|
Evolution of a distinct genomic domain in Drosophila: comparative analysis of the dot chromosome in Drosophila melanogaster and Drosophila virilis. Genetics 2010; 185:1519-34. [PMID: 20479145 DOI: 10.1534/genetics.110.116129] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The distal arm of the fourth ("dot") chromosome of Drosophila melanogaster is unusual in that it exhibits an amalgamation of heterochromatic properties (e.g., dense packaging, late replication) and euchromatic properties (e.g., gene density similar to euchromatic domains, replication during polytenization). To examine the evolution of this unusual domain, we undertook a comparative study by generating high-quality sequence data and manually curating gene models for the dot chromosome of D. virilis (Tucson strain 15010-1051.88). Our analysis shows that the dot chromosomes of D. melanogaster and D. virilis have higher repeat density, larger gene size, lower codon bias, and a higher rate of gene rearrangement compared to a reference euchromatic domain. Analysis of eight "wanderer" genes (present in a euchromatic chromosome arm in one species and on the dot chromosome in the other) shows that their characteristics are similar to other genes in the same domain, which suggests that these characteristics are features of the domain and are not required for these genes to function. Comparison of this strain of D. virilis with the strain sequenced by the Drosophila 12 Genomes Consortium (Tucson strain 15010-1051.87) indicates that most genes on the dot are under weak purifying selection. Collectively, despite the heterochromatin-like properties of this domain, genes on the dot evolve to maintain function while being responsive to changes in their local environment.
Collapse
|
34
|
Essential loci in centromeric heterochromatin of Drosophila melanogaster. I: the right arm of chromosome 2. Genetics 2010; 185:479-95. [PMID: 20382826 DOI: 10.1534/genetics.110.117259] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
With the most recent releases of the Drosophila melanogaster genome sequences, much of the previously absent heterochromatic sequences have now been annotated. We undertook an extensive genetic analysis of existing lethal mutations, as well as molecular mapping and sequence analysis (using a candidate gene approach) to identify as many essential genes as possible in the centromeric heterochromatin on the right arm of the second chromosome (2Rh) of D. melanogaster. We also utilized available RNA interference lines to knock down the expression of genes in 2Rh as another approach to identifying essential genes. In total, we verified the existence of eight novel essential loci in 2Rh: CG17665, CG17683, CG17684, CG17883, CG40127, CG41265, CG42595, and Atf6. Two of these essential loci, CG41265 and CG42595, are synonymous with the previously characterized loci l(2)41Ab and unextended, respectively. The genetic and molecular analysis of the previously reported locus, l(2)41Ae, revealed that this is not a single locus, but rather it is a large region of 2Rh that extends from unextended (CG42595) to CG17665 and includes four of the novel loci uncovered here.
Collapse
|
35
|
Grushko OG, Sharakhova MV, Stegnii VN, Sharakhov IV. Molecular organization of heterochromatin in malaria mosquitoes of the Anopheles maculipennis subgroup. Gene 2009; 448:192-7. [DOI: 10.1016/j.gene.2009.07.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 07/17/2009] [Accepted: 07/24/2009] [Indexed: 10/20/2022]
|
36
|
Abstract
The Y chromosome of Drosophila melanogaster has <20 protein-coding genes. These genes originated from the duplication of autosomal genes and have male-related functions. In 1993, Russell and Kaiser found three Y-linked pseudogenes of the Mst77F gene, which is a testis-expressed autosomal gene that is essential for male fertility. We did a thorough search using experimental and computational methods and found 18 Y-linked copies of this gene (named Mst77Y-1-Mst77Y-18). Ten Mst77Y genes encode defective proteins and the other eight are potentially functional. These eight genes produce approximately 20% of the functional Mst77F-like mRNA, and molecular evolutionary analysis shows that they evolved under purifying selection. Hence several Mst77Y genes have all the features of functional genes. Mst77Y genes are present only in D. melanogaster, and phylogenetic analysis confirmed that the duplication is a recent event. The identification of functional Mst77Y genes reinforces the previous finding that gene gains play a prominent role in the evolution of the Drosophila Y chromosome.
Collapse
|
37
|
Diverse roles of HP1 proteins in heterochromatin assembly and functions in fission yeast. Proc Natl Acad Sci U S A 2009; 106:8998-9003. [PMID: 19443688 DOI: 10.1073/pnas.0813063106] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Conserved chromosomal HP1 proteins capable of binding to histone H3 methylated at lysine 9 are believed to provide a dynamic platform for the recruitment and/or spreading of various regulatory proteins involved in diverse chromosomal processes. The fission yeast Schizosaccharomyces pombe HP1 family members Chp2 and Swi6 are important for heterochromatin assembly and transcriptional silencing, but their precise roles are not fully understood. Here, we show that Swi6 and Chp2 associate with histone deacetylase (HDAC) protein complexes containing class I HDAC Clr6 and class II HDAC Clr3 (a component of Snf2/HDAC repressor complex), which are critical for transcriptional silencing of centromeric repeats targeted by the heterochromatin machinery. Mapping of RNA polymerase (Pol) II distribution in single and double mutant backgrounds revealed that Swi6 and Chp2 proteins and their associated HDAC complexes have overlapping functions in limiting Pol II occupancy across pericentromeric heterochromatin domains. The purified Swi6 fraction also contains factors involved in various chromosomal processes such as chromatin remodeling and DNA replication. Also, Swi6 copurifies with Mis4 protein, a cohesin loading factor essential for sister chromatid cohesion, and with centromere-specific histone H3 variant CENP-A, which is incorporated into chromatin in a heterochromatin-dependent manner. These analyses suggest that among other functions, HP1 proteins associate with chromatin-modifying factors that in turn cooperate to assemble repressive chromatin; thus, precluding accessibility of underlying DNA sequences to transcriptional machinery.
Collapse
|
38
|
Dimitri P, Caizzi R, Giordano E, Carmela Accardo M, Lattanzi G, Biamonti G. Constitutive heterochromatin: a surprising variety of expressed sequences. Chromosoma 2009; 118:419-35. [PMID: 19412619 DOI: 10.1007/s00412-009-0211-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Revised: 03/30/2009] [Accepted: 04/01/2009] [Indexed: 10/20/2022]
Abstract
The organization of chromosomes into euchromatin and heterochromatin is amongst the most important and enigmatic aspects of genome evolution. Constitutive heterochromatin is a basic yet still poorly understood component of eukaryotic chromosomes, and its molecular characterization by means of standard genomic approaches is intrinsically difficult. Although recent evidence indicates that the presence of transcribed genes in constitutive heterochromatin is a conserved trait that accompanies the evolution of eukaryotic genomes, the term heterochromatin is still considered by many as synonymous of gene silencing. In this paper, we comprehensively review data that provide a clearer picture of transcribed sequences within constitutive heterochromatin, with a special emphasis on Drosophila and humans.
Collapse
Affiliation(s)
- Patrizio Dimitri
- Laboratorio di Genomica Funzionale e Proteomica di Sistemi modello and Istituto Pasteur-Fondazione Bolognetti, Dipartimento di Genetica e Biologia Molecolare Charles Darwin, Università La Sapienza, 00185, Italy.
| | | | | | | | | | | |
Collapse
|
39
|
Schaeffer SW, Bhutkar A, McAllister BF, Matsuda M, Matzkin LM, O'Grady PM, Rohde C, Valente VLS, Aguadé M, Anderson WW, Edwards K, Garcia ACL, Goodman J, Hartigan J, Kataoka E, Lapoint RT, Lozovsky ER, Machado CA, Noor MAF, Papaceit M, Reed LK, Richards S, Rieger TT, Russo SM, Sato H, Segarra C, Smith DR, Smith TF, Strelets V, Tobari YN, Tomimura Y, Wasserman M, Watts T, Wilson R, Yoshida K, Markow TA, Gelbart WM, Kaufman TC. Polytene chromosomal maps of 11 Drosophila species: the order of genomic scaffolds inferred from genetic and physical maps. Genetics 2008; 179:1601-55. [PMID: 18622037 PMCID: PMC2475758 DOI: 10.1534/genetics.107.086074] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2007] [Accepted: 03/13/2008] [Indexed: 11/18/2022] Open
Abstract
The sequencing of the 12 genomes of members of the genus Drosophila was taken as an opportunity to reevaluate the genetic and physical maps for 11 of the species, in part to aid in the mapping of assembled scaffolds. Here, we present an overview of the importance of cytogenetic maps to Drosophila biology and to the concepts of chromosomal evolution. Physical and genetic markers were used to anchor the genome assembly scaffolds to the polytene chromosomal maps for each species. In addition, a computational approach was used to anchor smaller scaffolds on the basis of the analysis of syntenic blocks. We present the chromosomal map data from each of the 11 sequenced non-Drosophila melanogaster species as a series of sections. Each section reviews the history of the polytene chromosome maps for each species, presents the new polytene chromosome maps, and anchors the genomic scaffolds to the cytological maps using genetic and physical markers. The mapping data agree with Muller's idea that the majority of Drosophila genes are syntenic. Despite the conservation of genes within homologous chromosome arms across species, the karyotypes of these species have changed through the fusion of chromosomal arms followed by subsequent rearrangement events.
Collapse
Affiliation(s)
- Stephen W Schaeffer
- Department of Biology and Institute of Molecular Evolutionary Genetics, The Pennsylvania State University, University Park, Pennsylvania 16802, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Chen ST, Cheng HC, Barbash DA, Yang HP. Evolution of hydra, a recently evolved testis-expressed gene with nine alternative first exons in Drosophila melanogaster. PLoS Genet 2008; 3:e107. [PMID: 17616977 PMCID: PMC1904467 DOI: 10.1371/journal.pgen.0030107] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Accepted: 05/15/2007] [Indexed: 12/26/2022] Open
Abstract
We describe here the Drosophila gene hydra that appears to have originated de novo in the melanogaster subgroup and subsequently evolved in both structure and expression level in Drosophila melanogaster and its sibling species. D. melanogaster hydra encodes a predicted protein of ~300 amino acids with no apparent similarity to any previously known proteins. The syntenic region flanking hydra on both sides is found in both D. ananassae and D. pseudoobscura, but hydra is found only in melanogaster subgroup species, suggesting that it originated less than ~13 million y ago. Exon 1 of hydra has undergone recurrent duplications, leading to the formation of nine tandem alternative exon 1s in D. melanogaster. Seven of these alternative exons are flanked on their 3′ side by the transposon DINE-1 (Drosophila interspersed element-1). We demonstrate that at least four of the nine duplicated exon 1s can function as alternative transcription start sites. The entire hydra locus has also duplicated in D. simulans and D. sechellia. D. melanogaster hydra is expressed most intensely in the proximal testis, suggesting a role in late-stage spermatogenesis. The coding region of hydra has a relatively high Ka/Ks ratio between species, but the ratio is less than 1 in all comparisons, suggesting that hydra is subject to functional constraint. Analysis of sequence polymorphism and divergence of hydra shows that it has evolved under positive selection in the lineage leading to D. melanogaster. The dramatic structural changes surrounding the first exons do not affect the tissue specificity of gene expression: hydra is expressed predominantly in the testes in D. melanogaster, D. simulans, and D. yakuba. However, we have found that expression level changed dramatically (~ >20-fold) between D. melanogaster and D. simulans. While hydra initially evolved in the absence of nearby transposable element insertions, we suggest that the subsequent accumulation of repetitive sequences in the hydra region may have contributed to structural and expression-level evolution by inducing rearrangements and causing local heterochromatinization. Our analysis further shows that recurrent evolution of both gene structure and expression level may be characteristics of newly evolved genes. We also suggest that late-stage spermatogenesis is the functional target for newly evolved and rapidly evolving male-specific genes. Similar groups of animals have similar numbers of genes, but not all of these genes are the same. While some genes are highly conserved and can be easily and uniquely identified in species ranging from yeast to plants to humans, other genes are sometimes found in only a small number or even in a single species. Such newly evolved genes may help produce traits that make species unique. We describe here a newly evolved gene called hydra that occurs only in a small subgroup of Drosophila species. hydra is expressed in the testes, suggesting that it may have a function in male fertility. hydra has evolved significantly in its structure and protein-coding sequence among species. The authors named the gene hydra after the nine-headed monster slain by Hercules because in one species, Drosophila melanogaster, hydra has nine potential alternative first exons. Perhaps because of this or other structural changes, the level of RNA made by hydra differs significantly between one pair of species. This analysis reveals that newly created genes may evolve rapidly in sequence, structure, and expression level.
Collapse
Affiliation(s)
- Shou-Tao Chen
- Faculty of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Hsin-Chien Cheng
- Faculty of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Daniel A Barbash
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Hsiao-Pei Yang
- Faculty of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan, Republic of China
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
41
|
Epigenetic regulation of heterochromatic DNA stability. Curr Opin Genet Dev 2008; 18:204-11. [PMID: 18372168 DOI: 10.1016/j.gde.2008.01.021] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Accepted: 01/16/2008] [Indexed: 02/08/2023]
Abstract
In this review we summarize recent studies that demonstrate the importance of epigenetic mechanisms for maintaining genome integrity, specifically with respect to repeated DNAs within heterochromatin. Potential problems that arise during replication, recombination, and repair of repeated sequences are counteracted by post-translational histone modifications and associated proteins, including the cohesins. These factors appear to ensure repeat stability by multiple mechanisms: suppressing homologous recombination, controlling the three-dimensional organization of damaged repeats to reduce the probability of aberrant recombination, and promoting the use of less problematic repair pathways. The presence of such systems may facilitate repeat and chromosome evolution, and their failure can lead to genome instability, chromosome rearrangements, and the onset of pathogenesis.
Collapse
|
42
|
Pezer Z, Ugarković D. RNA Pol II promotes transcription of centromeric satellite DNA in beetles. PLoS One 2008; 3:e1594. [PMID: 18270581 PMCID: PMC2220036 DOI: 10.1371/journal.pone.0001594] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2007] [Accepted: 01/17/2008] [Indexed: 12/04/2022] Open
Abstract
Transcripts of centromeric satellite DNAs are known to play a role in heterochromatin formation as well as in establishment of the kinetochore. However, little is known about basic mechanisms of satellite DNA expression within constitutive heterochromatin and its regulation. Here we present comprehensive analysis of transcription of abundant centromeric satellite DNA, PRAT from beetle Palorus ratzeburgii (Coleoptera). This satellite is characterized by preservation and extreme sequence conservation among evolutionarily distant insect species. PRAT is expressed in all three developmental stages: larvae, pupae and adults at similar level. Transcripts are abundant comprising 0.033% of total RNA and are heterogeneous in size ranging from 0.5 kb up to more than 5 kb. Transcription proceeds from both strands but with 10 fold different expression intensity and transcripts are not processed into siRNAs. Most of the transcripts (80%) are not polyadenylated and remain in the nucleus while a small portion is exported to the cytoplasm. Multiple, irregularly distributed transcription initiation sites as well as termination sites have been mapped within the PRAT sequence using primer extension and RLM-RACE. The presence of cap structure as well as poly(A) tails in a portion of the transcripts indicate RNA polymerase II–dependent transcription and a putative polymerase II promoter site overlaps the most conserved part of the PRAT sequence. The treatment of larvae with alpha-amanitin decreases the level of PRAT transcripts at concentrations that selectively inhibit pol II activity. In conclusion, stable, RNA polymerase II dependant transcripts of abundant centromeric satellite DNA, not regulated by RNAi, have been identified and characterized. This study offers a basic understanding of expression of highly abundant heterochromatic DNA which in beetle species constitutes up to 50% of the genome.
Collapse
Affiliation(s)
- Zeljka Pezer
- Department of Molecular Biology, Ruder Bosković Institute, Zagreb, Croatia
| | | |
Collapse
|
43
|
Yasuhara JC, Wakimoto BT. Molecular landscape of modified histones in Drosophila heterochromatic genes and euchromatin-heterochromatin transition zones. PLoS Genet 2007; 4:e16. [PMID: 18208336 PMCID: PMC2211541 DOI: 10.1371/journal.pgen.0040016] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2007] [Accepted: 12/10/2007] [Indexed: 01/11/2023] Open
Abstract
Constitutive heterochromatin is enriched in repetitive sequences and histone H3-methylated-at-lysine 9. Both components contribute to heterochromatin's ability to silence euchromatic genes. However, heterochromatin also harbors hundreds of expressed genes in organisms such as Drosophila. Recent studies have provided a detailed picture of sequence organization of D. melanogaster heterochromatin, but how histone modifications are associated with heterochromatic sequences at high resolution has not been described. Here, distributions of modified histones in the vicinity of heterochromatic genes of normal embryos and embryos homozygous for a chromosome rearrangement were characterized using chromatin immunoprecipitation and genome tiling arrays. We found that H3-di-methylated-at-lysine 9 (H3K9me2) was depleted at the 5′ ends but enriched throughout transcribed regions of heterochromatic genes. The profile was distinct from that of euchromatic genes and suggests that heterochromatic genes are integrated into, rather than insulated from, the H3K9me2-enriched domain. Moreover, the profile was only subtly affected by a Su(var)3–9 null mutation, implicating a histone methyltransferase other than SU(VAR)3–9 as responsible for most H3K9me2 associated with heterochromatic genes in embryos. On a chromosomal scale, we observed a sharp transition to the H3K9me2 domain, which coincided with increased retrotransposon density in the euchromatin-heterochromatin (eu-het) transition zones on the long chromosome arms. Thus, a certain density of retrotransposons, rather than specific boundary elements, may demarcate Drosophila pericentric heterochromatin. We also demonstrate that a chromosome rearrangement that created a new eu-het junction altered H3K9me2 distribution and induced new euchromatic sites of enrichment as far as several megabases away from the breakpoint. Taken together, the findings argue against simple classification of H3K9me as the definitive signature of silenced genes, and clarify roles of histone modifications and repetitive DNAs in heterochromatin. The results are also relevant for understanding the effects of chromosome aberrations and the megabase scale over which epigenetic position effects can operate in multicellular organisms. The chromosomal domain “heterochromatin” was first defined at the cytological level by its deeply staining appearance compared to more lightly stained domains called “euchromatin.” Abnormal juxtaposition of these two domains by chromosome rearrangements results in silencing of the nearby euchromatic genes. This effect is mediated by heterochromatin-enriched chromosomal proteins and led to the prevalent view of heterochromatin as incompatible with gene expression. Paradoxically, some expressed genes reside within heterochromatin. In this study, we examined how heterochromatic genes fit into a genomic context known for silencing effects. We found that Drosophila heterochromatic genes are integrated into the domain enriched in the modified histone H3K9me2, suggesting that the effect of this protein on gene expression is context-dependent. We also investigated the molecular nature of euchromatin-heterochromatin transition zones in the normal and rearranged chromosomes. The results provide insights into the functions of repetitive DNAs and H3K9me2 in heterochromatin and document the long distance over which a heterochromatic breakpoint can affect the molecular landscape of a chromosomal region. These findings have implications for understanding the consequences of chromosome abnormalities in organisms, including humans.
Collapse
Affiliation(s)
- Jiro C Yasuhara
- Department of Biology, University of Washington, Seattle, Washington, United States of America
| | - Barbara T Wakimoto
- Department of Biology, University of Washington, Seattle, Washington, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
44
|
Chaouki AS, Salz HK. Drosophila SPF45: a bifunctional protein with roles in both splicing and DNA repair. PLoS Genet 2007; 2:e178. [PMID: 17154718 PMCID: PMC1687153 DOI: 10.1371/journal.pgen.0020178] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2006] [Accepted: 09/01/2006] [Indexed: 11/18/2022] Open
Abstract
The sequence of the SPF45 protein is significantly conserved, yet functional studies have identified it as a splicing factor in animal cells and as a DNA-repair protein in plants. Using a combined genetic and biochemical approach to investigate this apparent functional discrepancy, we unify and validate both of these studies by demonstrating that the Drosophila melanogaster protein is bifunctional, with independent functions in DNA repair and splicing. We find that SPF45 associates with the U2 snRNP and that mutations that remove the C-terminal end of the protein disrupt this interaction. Although animals carrying this mutation are viable, they are nevertheless compromised in their ability to regulate Sex-lethal splicing, demonstrating that Sex-lethal is an important physiological target of SPF45. Furthermore, these mutant animals exhibit phenotypes diagnostic of difficulties in recovering from exogenously induced DNA damage. The conclusion that SPF45 functions in the DNA-repair pathway is strengthened by finding both genetic and physical interactions between SPF45 and RAD201, a previously uncharacterized member of the RecA/Rad51 protein family. Together with our finding that the fly SPF45 protein increases the survival rate of mutagen-treated bacteria lacking the RecG helicase, these studies provide the tantalizing suggestion that SPF45 has an ancient and evolutionarily conserved role in DNA repair. Assigning function to a protein relies on information about similar proteins in different species, and is based on the view that conservation of sequence generally parallels conservation of function. In this article, Chaouki and Salz focus on SPF45, a protein that, at first glance, appears to break this rule. Although the sequence of SPF45 is highly conserved, in animals cells SPF45 functions as a splicing factor, but in plant cells it functions as a DNA repair protein. This functional discrepancy is resolved here through the demonstration that, in D. melanogaster, SPF45 is a bifunctional protein with independent functions in DNA repair and splicing. Support for this conclusion includes the observation that mutant animals lacking SPF45 function display defects in both splicing and DNA repair. In addition, the authors show that SPF45 associates with two distinct groups of proteins; those that participate in RNA splicing and those that participate in DNA repair. The finding that the D. melanogaster protein is bifunctional suggests that the human protein may also have more than one function. This has important clinical implications because elevated SPF45 levels have been correlated with resistance to chemotherapy.
Collapse
Affiliation(s)
- Ahmad Sami Chaouki
- Department of Genetics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Helen K Salz
- Department of Genetics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
45
|
Abstract
Rice (Oryza sativa) has become an important model plant species in numerous research projects involving genome, molecular and evolutionary biology. In this review we describe the reasons why rice provides an excellent model system for centromere and heterochromatin research. In most multicellular eukaryotes, centromeres and heterochromatic domains contain long arrays of repetitive DNA elements that are recalcitrant to DNA sequencing. In contrast, three rice centromeres and the majority of the cytologically defined heterochromatin in the rice genome have been sequenced to high quality, providing an unparalleled resource compared to other model multicellular eukaryotes. Most importantly, active genes have been discovered in the functional domains of several rice centromeres. The centromeric genes and sequence resources provide an unprecedented opportunity to study function and evolution of centromeres and centromere-associated genes.
Collapse
Affiliation(s)
- Huihuang Yan
- Department of Horticulture, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | |
Collapse
|
46
|
Complex regulation and multiple developmental functions of misfire, the Drosophila melanogaster ferlin gene. BMC DEVELOPMENTAL BIOLOGY 2007; 7:21. [PMID: 17386097 PMCID: PMC1853072 DOI: 10.1186/1471-213x-7-21] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2006] [Accepted: 03/26/2007] [Indexed: 11/30/2022]
Abstract
Background Ferlins are membrane proteins with multiple C2 domains and proposed functions in Ca2+ mediated membrane-membrane interactions in animals. Caenorhabditis elegans has two ferlin genes, one of which is required for sperm function. Mammals have several ferlin genes and mutations in the human dysferlin (DYSF) and otoferlin (OTOF) genes result in muscular dystrophy and hearing loss, respectively. Drosophila melanogaster has a single ferlin gene called misfire (mfr). A previous study showed that a mfr mutation caused male sterility because of defects in fertilization. Here we analyze the expression and structure of the mfr gene and the consequences of multiple mutations to better understand the developmental function of ferlins. Results We show that mfr is expressed in the testis and ovaries of adult flies, has tissue-specific promoters, and expresses alternatively spliced transcripts that are predicted to encode distinct protein isoforms. Studies of 11 male sterile mutations indicate that a predicted Mfr testis isoform with five C2 domains and a transmembrane (TM) domain is required for sperm plasma membrane breakdown (PMBD) and completion of sperm activation during fertilization. We demonstrate that Mfr is not required for localization of Sneaky, another membrane protein necessary for PMBD. The mfr mutations vary in their effects in females, with a subset disrupting egg patterning and causing a maternal effect delay in early embryonic development. Locations of these mutations indicate that a short Mfr protein isoform carries out ferlin activities during oogenesis. Conclusion The mfr gene exhibits complex transcriptional and post-transcriptional regulation and functions in three developmental processes: sperm activation, egg patterning, and early embryogenesis. These functions are in part due to the production of protein isoforms that vary in the number of C2 domains. These findings help establish D. melanogaster as model system for understanding ferlin function and dysfunction in animals, including humans.
Collapse
|
47
|
Gvozdev VA, Abramov YA, Kogan GL, Lavrov SA. Distorted heterochromatin replication in Drosophila melanogaster polytene chromosomes as a result of euchromatin-heterochromatin rearrangements. RUSS J GENET+ 2007. [DOI: 10.1134/s1022795407010024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
48
|
Schulze SR, Wallrath LL. Gene regulation by chromatin structure: paradigms established in Drosophila melanogaster. ANNUAL REVIEW OF ENTOMOLOGY 2007; 52:171-92. [PMID: 16881818 DOI: 10.1146/annurev.ento.51.110104.151007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Studies in Drosophila melanogaster have revealed paradigms for regulating gene expression through chromatin structure, including mechanisms of gene activation and silencing. Regulation occurs at the level of individual genes, chromosomal domains, and entire chromosomes. The chromatin state is dynamic, allowing for changes in gene expression in response to cellular signals and/or environmental cues. Changes in chromatin result from the action of ATP-dependent chromatin-remodeling complexes, reversible epigenetic histone modifications, and the incorporation of histone variants. Many of the chromatin-based transcriptional regulatory mechanisms discovered in D. melanogaster are evolutionarily conserved and therefore serve as a foundation for studies in other organisms.
Collapse
Affiliation(s)
- Sandra R Schulze
- Department of Biology, Western Washington University, Bellingham, Washington 98225, USA.
| | | |
Collapse
|
49
|
Fan C, Long M. A New Retroposed Gene in Drosophila Heterochromatin Detected by Microarray-Based Comparative Genomic Hybridization. J Mol Evol 2006; 64:272-83. [PMID: 17177089 DOI: 10.1007/s00239-006-0169-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2006] [Accepted: 08/17/2006] [Indexed: 10/23/2022]
Abstract
A genomic pattern of new gene origination is often dependent on a genomic method that can efficiently identify a statistically adequate number of recently originated genes. The heterochromatic regions have often been viewed as genomic deserts with low coding potential and thus a low flux of new genes. However, increasing reports revealed unexpected roles of heterochromatic regions in the evolution of genes and genomes. We identified recently retroposed genes that originated in heterochromatic regions in Drosophila, by developing microarray-based comparative genomic hybridization (CGH) with multiple species. This new gene family, named Ifc-2h, originated in the common ancestor of the clade of D. simulans, D. mauritiana, and D. sechellia. The sequence features and phylogenetic distribution indicated that Ifc-2h resulted from the retroposition from its parental gene, Infertile crescent (Ifc), and integrated into heterochromatic region of common ancestor of the three sibling species 2 million years ago. Expression analysis revealed that Ifc-2h had developed a new expression pattern by recruiting a putative regulatory element from its target sequence. The distribution of indel variation in Ifc-2h of D. simulans and D. mauritiana revealed a significant sequence constraint, suggesting that the Ifc-2h gene may be functional. These analyses cast fresh insight into the evolution of heterochromatin and the origin of its coding regions.
Collapse
Affiliation(s)
- Chuanzhu Fan
- Department of Ecology and Evolution, The University of Chicago, 1101 East 57th Street, Chicago, IL 60637, USA
| | | |
Collapse
|
50
|
Rossi F, Moschetti R, Caizzi R, Corradini N, Dimitri P. Cytogenetic and molecular characterization of heterochromatin gene models in Drosophila melanogaster. Genetics 2006; 175:595-607. [PMID: 17110485 PMCID: PMC1800633 DOI: 10.1534/genetics.106.065441] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In the past decade, genome-sequencing projects have yielded a great amount of information on DNA sequences in several organisms. The release of the Drosophila melanogaster heterochromatin sequence by the Drosophila Heterochromatin Genome Project (DHGP) has greatly facilitated studies of mapping, molecular organization, and function of genes located in pericentromeric heterochromatin. Surprisingly, genome annotation has predicted at least 450 heterochromatic gene models, a figure 10-fold above that defined by genetic analysis. To gain further insight into the locations and functions of D. melanogaster heterochromatic genes and genome organization, we have FISH mapped 41 gene models relative to the stained bands of mitotic chromosomes and the proximal divisions of polytene chromosomes. These genes are contained in eight large scaffolds, which together account for approximately 1.4 Mb of heterochromatic DNA sequence. Moreover, developmental Northern analysis showed that the expression of 15 heterochromatic gene models tested is similar to that of the vital heterochromatic gene Nipped-A, in that it is not limited to specific stages, but is present throughout all development, despite its location in a supposedly "silent" region of the genome. This result is consistent with the idea that genes resident in heterochromatin can encode essential functions.
Collapse
Affiliation(s)
- Fabrizio Rossi
- Laboratorio di Genomica Funzionale e Proteomica di Sistemi complessi, Dipartimento di Genetica e Biologia Molecolare Charles Darwin, Università La Sapienza, 00185 Roma, Italy
| | | | | | | | | |
Collapse
|