1
|
Newman T, Bond DM, Ishihara T, Rizzoli P, Gouil Q, Hore TA, Shaw G, Renfree MB. PRKACB is a novel imprinted gene in marsupials. Epigenetics Chromatin 2024; 17:29. [PMID: 39342354 PMCID: PMC11438212 DOI: 10.1186/s13072-024-00552-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/22/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Genomic imprinting results in parent-of-origin-specific gene expression and, among vertebrates, is found only in therian mammals: marsupials and eutherians. A differentially methylated region (DMR), in which the methylation status of CpG dinucleotides differs between the two alleles, can mark the parental identity of imprinted genes. We developed a computational pipeline that detected CpG islands (CGIs) marked by both methylated and unmethylated signals in whole genome bisulfite sequencing data. This approach identified candidate marsupial DMRs in a publicly available koala methylome. One of these candidate DMRs was associated with PRKACB, a gene encoding the protein kinase A catalytic subunit beta. Nothing is known about the imprinting status of PRKACB in eutherian mammals although mutations of this gene are associated with endocrine neoplasia and other developmental disorders. RESULTS In the tammar wallaby and brushtail possum there was parent-of-origin-specific DNA methylation in the PRKACB DMR in which the maternal allele was methylated and the paternal allele was unmethylated. There were multiple RNAs transcribed from this locus. Allele-specific expression analysis identified paternal expression of a PRKACB lncRNA and an mRNA isoform. Comparison of the PRKACB gene start site between marsupials and eutherians demonstrated that the CGI is longer in marsupials. The PRKACB gene product functions in the same signalling pathway as the guanine nucleotide-binding protein alpha subunit encoded at the GNAS locus, a known eutherian imprinted gene. In a mouse methylome Gnas had three differentially methylated CGIs, while in the koala methylome the GNAS locus had two unmethylated CGIs. CONCLUSIONS We conclude that PRKACB is a novel, DMR-associated marsupial imprinted gene. Imprinting of PRKACB in marsupials and GNAS in eutherians may indicate a conserved selection pressure for imprinting of the protein kinase A signalling pathway in therians with the two lineages adapting by imprinting different genes.
Collapse
Affiliation(s)
- Trent Newman
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Donna M Bond
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Teruhito Ishihara
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Epigenetics Programme, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Phoebe Rizzoli
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Quentin Gouil
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, 3010, Australia
| | - Timothy A Hore
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Geoff Shaw
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
2
|
Herbert A. Neurodegenerative diseases reflect the reciprocal roles played by retroelements in regulating memory and immunity. Front Neurosci 2024; 18:1445540. [PMID: 39371608 PMCID: PMC11451048 DOI: 10.3389/fnins.2024.1445540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Tetrapod endogenous retroelements (ERE) encode proteins that have been exapted to perform many roles in development and also in innate immunity, including GAG (group specific antigen) proteins from the ERE long terminal repeat (LTR) family, some of which can assemble into viral-like capsids (VLCs) and transmit mRNA across synapses. The best characterized member of this family is ARC (activity-regulated cytoskeletal gene), that is involved in memory formation. Other types of EREs, such as LINES and SINES (long and short interspersed elements), have instead been exapted for immune defenses against infectious agents. These immune EREs identify host transcripts by forming the unusual left-handed Z-DNA and Z-RNA conformations to enable self/nonself discrimination. Elevated levels of immune EREs in the brain are associated with neurodegenerative disease. Here I address the question of how pathways based on immune EREs are relate to the memory EREs that mediate neural plasticity. I propose that during infection and in other inflammatory states, ERE encoded GAG capsids deliver interferon-induced immune EREs that rapidly inhibit translation of viral RNAs in the dendritic splines by activation of protein kinase R (PKR). The response limits transmission of viruses and autonomously replicating elements, while protecting bystander cells from stress-induced cell death. Further, the PKR-dependent phosphorylation of proteins, like tau, disrupts the endocytic pathways exploited by viruses to spread to other cells. The responses come at a cost. They impair memory formation and can contribute to pathology by increasing the deposition of amyloid beta.
Collapse
|
3
|
Del Toro-De León G, van Boven J, Santos-González J, Jiao WB, Peng H, Schneeberger K, Köhler C. Epigenetic and transcriptional consequences in the endosperm of chemically induced transposon mobilization in Arabidopsis. Nucleic Acids Res 2024; 52:8833-8848. [PMID: 38967011 PMCID: PMC11347142 DOI: 10.1093/nar/gkae572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/13/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024] Open
Abstract
Genomic imprinting, an epigenetic phenomenon leading to parent-of-origin-specific gene expression, has independently evolved in the endosperm of flowering plants and the placenta of mammals-tissues crucial for nurturing embryos. While transposable elements (TEs) frequently colocalize with imprinted genes and are implicated in imprinting establishment, direct investigations of the impact of de novo TE transposition on genomic imprinting remain scarce. In this study, we explored the effects of chemically induced transposition of the Copia element ONSEN on genomic imprinting in Arabidopsis thaliana. Through the combination of chemical TE mobilization and doubled haploid induction, we generated a line with 40 new ONSEN copies. Our findings reveal a preferential targeting of maternally expressed genes (MEGs) for transposition, aligning with the colocalization of H2A.Z and H3K27me3 in MEGs-both previously identified as promoters of ONSEN insertions. Additionally, we demonstrate that chemically-induced DNA hypomethylation induces global transcriptional deregulation in the endosperm, leading to the breakdown of MEG imprinting. This study provides insights into the consequences of chemically induced TE remobilization in the endosperm, revealing that chemically-induced epigenome changes can have long-term consequences on imprinted gene expression.
Collapse
Affiliation(s)
- Gerardo Del Toro-De León
- Department of Plant Reproductive Biology and Epigenetics, Max Planck Institute of Molecular Plant Physiology, Potsdam 14476, Germany
| | - Joram van Boven
- Department of Plant Biology, Uppsala BioCenter, Swedish University of Agricultural Sciences and Linnean Centre for Plant Biology, Uppsala 75007, Sweden
| | - Juan Santos-González
- Department of Plant Biology, Uppsala BioCenter, Swedish University of Agricultural Sciences and Linnean Centre for Plant Biology, Uppsala 75007, Sweden
| | - Wen-Biao Jiao
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, Huazhong Agricultural University, Wuhan 430070, China
| | - Haoran Peng
- Department of Plant Reproductive Biology and Epigenetics, Max Planck Institute of Molecular Plant Physiology, Potsdam 14476, Germany
| | - Korbinian Schneeberger
- Department of Chromosome Biology, Max Planck Institute for Plant Breeding Research, Cologne 50829, Germany
- Faculty for Biology, LMU Munich, Planegg-Martinsried 82152, Germany
- Cluster of Excellence on Plant Sciences, Heinrich-Heine University, Düsseldorf 40225, Germany
| | - Claudia Köhler
- Department of Plant Reproductive Biology and Epigenetics, Max Planck Institute of Molecular Plant Physiology, Potsdam 14476, Germany
- Department of Plant Biology, Uppsala BioCenter, Swedish University of Agricultural Sciences and Linnean Centre for Plant Biology, Uppsala 75007, Sweden
| |
Collapse
|
4
|
Newman T, Ishihara T, Shaw G, Renfree MB. The structure of the TH/INS locus and the parental allele expressed are not conserved between mammals. Heredity (Edinb) 2024; 133:21-32. [PMID: 38834866 PMCID: PMC11222543 DOI: 10.1038/s41437-024-00689-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 05/01/2024] [Accepted: 05/07/2024] [Indexed: 06/06/2024] Open
Abstract
Parent-of-origin-specific expression of imprinted genes is critical for successful mammalian growth and development. Insulin, coded by the INS gene, is an important growth factor expressed from the paternal allele in the yolk sac placenta of therian mammals. The tyrosine hydroxylase gene TH encodes an enzyme involved in dopamine synthesis. TH and INS are closely associated in most vertebrates, but the mouse orthologues, Th and Ins2, are separated by repeated DNA. In mice, Th is expressed from the maternal allele, but the parental origin of expression is not known for any other mammal so it is unclear whether the maternal expression observed in the mouse represents an evolutionary divergence or an ancestral condition. We compared the length of the DNA segment between TH and INS across species and show that separation of these genes occurred in the rodent lineage with an accumulation of repeated DNA. We found that the region containing TH and INS in the tammar wallaby produces at least five distinct RNA transcripts: TH, TH-INS1, TH-INS2, lncINS and INS. Using allele-specific expression analysis, we show that the TH/INS locus is expressed from the paternal allele in pre- and postnatal tammar wallaby tissues. Determining the imprinting pattern of TH/INS in other mammals might clarify if paternal expression is the ancestral condition which has been flipped to maternal expression in rodents by the accumulation of repeat sequences.
Collapse
Affiliation(s)
- Trent Newman
- School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Teruhito Ishihara
- School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
- Epigenetics Programme, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Geoff Shaw
- School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
Honda S, Hatamura M, Kunimoto Y, Ikeda S, Minami N. Chimeric PRMT6 protein produced by an endogenous retrovirus promoter regulates cell fate decision in mouse preimplantation embryos†. Biol Reprod 2024; 110:698-710. [PMID: 38196172 DOI: 10.1093/biolre/ioae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/11/2023] [Accepted: 01/07/2023] [Indexed: 01/11/2024] Open
Abstract
Murine endogenous retrovirus with leucine tRNA primer, also known as MERVL, is expressed during zygotic genome activation in mammalian embryos. Here we show that protein arginine N-methyltransferase 6 (Prmt6) forms a chimeric transcript with MT2B2, one of the long terminal repeat sequences of murine endogenous retrovirus with leucine tRNA primer, and is translated into an elongated chimeric protein (PRMT6MT2B2) whose function differs from that of the canonical PRMT6 protein (PRMT6CAN) in mouse preimplantation embryos. Overexpression of PRMT6CAN in fibroblast cells increased asymmetric dimethylation of the third arginine residue of both histone H2A (H2AR3me2a) and histone H4 (H4R3me2a), while overexpression of PRMT6MT2B2 increased only H2AR3me2a. In addition, overexpression of PRMT6MT2B2 in one blastomere of mouse two-cell embryos promoted cell proliferation and differentiation of the blastomere into epiblast cells at the blastocyst stage, while overexpression of PRMT6CAN repressed cell proliferation. This is the first report of the translation of a chimeric protein (PRMT6MT2B2) in mouse preimplantation embryos. Our results suggest that analyzing chimeric transcripts with murine endogenous retrovirus with leucine tRNA primer will provide insight into the relationship between zygotic genome activation and subsequent intra- and extra-cellular lineage determination.
Collapse
Affiliation(s)
- Shinnosuke Honda
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Maho Hatamura
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Yuri Kunimoto
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Shuntaro Ikeda
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Naojiro Minami
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| |
Collapse
|
6
|
Henriques WS, Young JM, Nemudryi A, Nemudraia A, Wiedenheft B, Malik HS. The Diverse Evolutionary Histories of Domesticated Metaviral Capsid Genes in Mammals. Mol Biol Evol 2024; 41:msae061. [PMID: 38507667 PMCID: PMC11011659 DOI: 10.1093/molbev/msae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024] Open
Abstract
Selfish genetic elements comprise significant fractions of mammalian genomes. In rare instances, host genomes domesticate segments of these elements for function. Using a complete human genome assembly and 25 additional vertebrate genomes, we re-analyzed the evolutionary trajectories and functional potential of capsid (CA) genes domesticated from Metaviridae, a lineage of retrovirus-like retrotransposons. Our study expands on previous analyses to unearth several new insights about the evolutionary histories of these ancient genes. We find that at least five independent domestication events occurred from diverse Metaviridae, giving rise to three universally retained single-copy genes evolving under purifying selection and two gene families unique to placental mammals, with multiple members showing evidence of rapid evolution. In the SIRH/RTL family, we find diverse amino-terminal domains, widespread loss of protein-coding capacity in RTL10 despite its retention in several mammalian lineages, and differential utilization of an ancient programmed ribosomal frameshift in RTL3 between the domesticated CA and protease domains. Our analyses also reveal that most members of the PNMA family in mammalian genomes encode a conserved putative amino-terminal RNA-binding domain (RBD) both adjoining and independent from domesticated CA domains. Our analyses lead to a significant correction of previous annotations of the essential CCDC8 gene. We show that this putative RBD is also present in several extant Metaviridae, revealing a novel protein domain configuration in retrotransposons. Collectively, our study reveals the divergent outcomes of multiple domestication events from diverse Metaviridae in the common ancestor of placental mammals.
Collapse
Affiliation(s)
- William S Henriques
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Janet M Young
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Artem Nemudryi
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Anna Nemudraia
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Blake Wiedenheft
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Harmit S Malik
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| |
Collapse
|
7
|
Genner R, Akeson S, Meredith M, Jerez PA, Malik L, Baker B, Miano-Burkhardt A, Paten B, Billingsley KJ, Blauwendraat C, Jain M. Assessing methylation detection for primary human tissue using Nanopore sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.581569. [PMID: 38464144 PMCID: PMC10925257 DOI: 10.1101/2024.02.29.581569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
DNA methylation most commonly occurs as 5-methylcytosine (5-mC) in the human genome and has been associated with human diseases. Recent developments in single-molecule sequencing technologies (Oxford Nanopore Technologies (ONT) and Pacific Biosciences) have enabled readouts of long, native DNA molecules, including cytosine methylation. ONT recently upgraded their Nanopore sequencing chemistry and kits from R9 to the R10 version, which yielded increased accuracy and sequencing throughput. However the effects on methylation detection have not yet been documented. Here we performed a series of computational analyses to characterize differences in Nanopore-based 5mC detection between the ONT R9 and R10 chemistries. We compared 5mC calls in R9 and R10 for three human genome datasets: a cell line, a frontal cortex brain sample, and a blood sample. We performed an in-depth analysis on CpG islands and homopolymer regions, and documented high concordance for methylation detection among sequencing technologies. The strongest correlation was observed between Nanopore R10 and Illumina bisulfite technologies for cell line-derived datasets. Subtle differences in methylation datasets between technologies can impact analysis tools such as differential methylation calling software. Our findings show that comparisons can be drawn between methylation data from different Nanopore chemistries using guided hypotheses. This work will facilitate comparison among Nanopore data cohorts derived using different chemistries from large scale sequencing efforts, such as the NIH CARD Long Read Initiative.
Collapse
Affiliation(s)
- Rylee Genner
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Stuart Akeson
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Melissa Meredith
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Pilar Alvarez Jerez
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Laksh Malik
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Breeana Baker
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Benedict Paten
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Kimberley J Billingsley
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Miten Jain
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Department of Bioengineering, Northeastern University, Boston, MA, USA
- Department of Physics, Northeastern University, Boston, MA, USA
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA
| |
Collapse
|
8
|
Kitazawa M. Evolution of the nervous system by acquisition of retrovirus-derived genes in mammals. Genes Genet Syst 2024; 98:321-336. [PMID: 38220159 DOI: 10.1266/ggs.23-00197] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
In the course of evolution, the most highly developed organ is likely the brain, which has become more complex over time and acquired diverse forms and functions in different species. In particular, mammals have developed complex and high-functioning brains, and it has been reported that several genes derived from retroviruses were involved in mammalian brain evolution, that is, generating the complexity of the nervous system. Especially, the sushi-ichi-related retrotransposon homolog (SIRH)/retrotransposon gag-like (RTL) genes have been suggested to play a role in the evolutionary processes shaping brain morphology and function in mammals. Genetic mutation and altered expression of genes are linked to neurological disorders, highlighting how the acquisition of virus-derived genes in mammals has both driven brain evolution and imposed a susceptibility to diseases. This review provides an overview of the functions, diversity, evolution and diseases associated with SIRH/RTL genes in the nervous system. The contribution of retroviruses to brain evolution is an important research topic in evolutionary biology and neuroscience, and further insights are expected to be gained through future studies.
Collapse
Affiliation(s)
- Moe Kitazawa
- School of BioSciences, Faculty of Science, The University of Melbourne
| |
Collapse
|
9
|
Ishihara T, Suzuki S, Newman TA, Fenelon JC, Griffith OW, Shaw G, Renfree MB. Marsupials have monoallelic MEST expression with a conserved antisense lncRNA but MEST is not imprinted. Heredity (Edinb) 2024; 132:5-17. [PMID: 37952041 PMCID: PMC10798977 DOI: 10.1038/s41437-023-00656-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 11/14/2023] Open
Abstract
The imprinted isoform of the Mest gene in mice is involved in key mammalian traits such as placental and fetal growth, maternal care and mammary gland maturation. The imprinted isoform has a distinct differentially methylated region (DMR) at its promoter in eutherian mammals but in marsupials, there are no differentially methylated CpG islands between the parental alleles. Here, we examined similarities and differences in the MEST gene locus across mammals using a marsupial, the tammar wallaby, a monotreme, the platypus, and a eutherian, the mouse, to investigate how imprinting of this gene evolved in mammals. By confirming the presence of the short isoform in all mammalian groups (which is imprinted in eutherians), this study suggests that an alternative promoter for the short isoform evolved at the MEST gene locus in the common ancestor of mammals. In the tammar, the short isoform of MEST shared the putative promoter CpG island with an antisense lncRNA previously identified in humans and an isoform of a neighbouring gene CEP41. The antisense lncRNA was expressed in tammar sperm, as seen in humans. This suggested that the conserved lncRNA might be important in the establishment of MEST imprinting in therian mammals, but it was not imprinted in the tammar. In contrast to previous studies, this study shows that MEST is not imprinted in marsupials. MEST imprinting in eutherians, therefore must have occurred after the marsupial-eutherian split with the acquisition of a key epigenetic imprinting control region, the differentially methylated CpG islands between the parental alleles.
Collapse
Affiliation(s)
- Teruhito Ishihara
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
| | - Shunsuke Suzuki
- Department of Agricultural and Life Sciences, Shinshu University, Nagano, Japan
| | - Trent A Newman
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Jane C Fenelon
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Oliver W Griffith
- Department of Biological Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Geoff Shaw
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
10
|
Bond DM, Ortega-Recalde O, Laird MK, Hayakawa T, Richardson KS, Reese FCB, Kyle B, McIsaac-Williams BE, Robertson BC, van Heezik Y, Adams AL, Chang WS, Haase B, Mountcastle J, Driller M, Collins J, Howe K, Go Y, Thibaud-Nissen F, Lister NC, Waters PD, Fedrigo O, Jarvis ED, Gemmell NJ, Alexander A, Hore TA. The admixed brushtail possum genome reveals invasion history in New Zealand and novel imprinted genes. Nat Commun 2023; 14:6364. [PMID: 37848431 PMCID: PMC10582058 DOI: 10.1038/s41467-023-41784-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 09/13/2023] [Indexed: 10/19/2023] Open
Abstract
Combining genome assembly with population and functional genomics can provide valuable insights to development and evolution, as well as tools for species management. Here, we present a chromosome-level genome assembly of the common brushtail possum (Trichosurus vulpecula), a model marsupial threatened in parts of their native range in Australia, but also a major introduced pest in New Zealand. Functional genomics reveals post-natal activation of chemosensory and metabolic genes, reflecting unique adaptations to altricial birth and delayed weaning, a hallmark of marsupial development. Nuclear and mitochondrial analyses trace New Zealand possums to distinct Australian subspecies, which have subsequently hybridised. This admixture allowed phasing of parental alleles genome-wide, ultimately revealing at least four genes with imprinted, parent-specific expression not yet detected in other species (MLH1, EPM2AIP1, UBP1 and GPX7). We find that reprogramming of possum germline imprints, and the wider epigenome, is similar to eutherian mammals except onset occurs after birth. Together, this work is useful for genetic-based control and conservation of possums, and contributes to understanding of the evolution of novel mammalian epigenetic traits.
Collapse
Affiliation(s)
- Donna M Bond
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | | | - Melanie K Laird
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Takashi Hayakawa
- Faculty of Environmental Earth Science, Hokkaido University, Sapporo, Hokkaido, 060-0808, Japan
| | - Kyle S Richardson
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Biology Department, University of Montana Western, Dillon, MT, 59725, USA
| | - Finlay C B Reese
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Bruce Kyle
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | | | | | | | - Amy L Adams
- Department of Zoology, University of Otago, Dunedin, New Zealand
| | - Wei-Shan Chang
- School of Life and Environmental Science, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
- Health and Biosecurity, CSIRO, Canberra, ACT, Australia
| | - Bettina Haase
- Vertebrate Genome Laboratory, The Rockefeller University, New York, NY, USA
| | | | | | - Joanna Collins
- Tree of Life, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Kerstin Howe
- Tree of Life, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Yasuhiro Go
- Graduate School of Information Science, Hyogo University, Hyogo, Japan
- Cognitive Genomics Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi, Japan
- Department of System Neuroscience, National Institute for Physiological Sciences, Aichi, Japan
| | - Francoise Thibaud-Nissen
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Nicholas C Lister
- School of Biotechnology and Biomolecular Science, Faculty of Science, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Paul D Waters
- School of Biotechnology and Biomolecular Science, Faculty of Science, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Olivier Fedrigo
- Vertebrate Genome Laboratory, The Rockefeller University, New York, NY, USA
| | - Erich D Jarvis
- Vertebrate Genome Laboratory, The Rockefeller University, New York, NY, USA
- Laboratory of Neurogenetics of Language, The Rockefeller University, New York, NY, 10065, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
| | - Neil J Gemmell
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Alana Alexander
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Timothy A Hore
- Department of Anatomy, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
11
|
Shimode S. Acquisition and Exaptation of Endogenous Retroviruses in Mammalian Placenta. Biomolecules 2023; 13:1482. [PMID: 37892164 PMCID: PMC10604696 DOI: 10.3390/biom13101482] [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: 08/15/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Endogenous retroviruses (ERVs) are retrovirus-like sequences that were previously integrated into the host genome. Although most ERVs are inactivated by mutations, deletions, or epigenetic regulation, some remain transcriptionally active and impact host physiology. Several ERV-encoded proteins, such as Syncytins and Suppressyn, contribute to placenta acquisition, a crucial adaptation in mammals that protects the fetus from external threats and other risks while enabling the maternal supply of oxygen, nutrients, and antibodies. In primates, Syncytin-1 and Syncytin-2 facilitate cell-cell fusion for placental formation. Suppressyn is the first ERV-derived protein that inhibits cell fusion by binding to ASCT2, the receptor for Syncytin-1. Furthermore, Syncytin-2 likely inserted into the genome of the common ancestor of Anthropoidea, whereas Syncytin-1 and Suppressyn likely inserted into the ancestor of catarrhines; however, they were inactivated in some lineages, suggesting that multiple exaptation events had occurred. This review discusses the role of ERV-encoded proteins, particularly Syncytins and Suppressyn, in placental development and function, focusing on the integration of ERVs into the host genome and their contribution to the genetic mechanisms underlying placentogenesis. This review provides valuable insights into the molecular and genetic aspects of placentation, potentially shedding light on broader evolutionary and physiological processes in mammals.
Collapse
Affiliation(s)
- Sayumi Shimode
- Genome Editing Innovation Center, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-0046, Japan;
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| |
Collapse
|
12
|
Ishino F, Itoh J, Irie M, Matsuzawa A, Naruse M, Suzuki T, Hiraoka Y, Kaneko-Ishino T. Retrovirus-Derived RTL9 Plays an Important Role in Innate Antifungal Immunity in the Eutherian Brain. Int J Mol Sci 2023; 24:14884. [PMID: 37834332 PMCID: PMC10573853 DOI: 10.3390/ijms241914884] [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: 08/16/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Retrotransposon Gag-like (RTL) genes play a variety of essential and important roles in the eutherian placenta and brain. It has recently been demonstrated that RTL5 and RTL6 (also known as sushi-ichi retrotransposon homolog 8 (SIRH8) and SIRH3) are microglial genes that play important roles in the brain's innate immunity against viruses and bacteria through their removal of double-stranded RNA and lipopolysaccharide, respectively. In this work, we addressed the function of RTL9 (also known as SIRH10). Using knock-in mice that produce RTL9-mCherry fusion protein, we examined RTL9 expression in the brain and its reaction to fungal zymosan. Here, we demonstrate that RTL9 plays an important role, degrading zymosan in the brain. The RTL9 protein is localized in the microglial lysosomes where incorporated zymosan is digested. Furthermore, in Rtl9 knockout mice expressing RTL9ΔC protein lacking the C-terminus retroviral GAG-like region, the zymosan degrading activity was lost. Thus, RTL9 is essentially engaged in this reaction, presumably via its GAG-like region. Together with our previous study, this result highlights the importance of three retrovirus-derived microglial RTL genes as eutherian-specific constituents of the current brain innate immune system: RTL9, RTL5 and RTL6, responding to fungi, viruses and bacteria, respectively.
Collapse
Affiliation(s)
- Fumitoshi Ishino
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (M.I.); (A.M.); (M.N.)
| | - Johbu Itoh
- Department of Pathology, School of Medicine, Tokai University, Isehara 259-1193, Japan;
| | - Masahito Irie
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (M.I.); (A.M.); (M.N.)
- Faculty of Nursing, School of Medicine, Tokai University, Isehara 259-1193, Japan
| | - Ayumi Matsuzawa
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (M.I.); (A.M.); (M.N.)
- Department of Genomic Function and Diversity, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Mie Naruse
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (M.I.); (A.M.); (M.N.)
| | - Toru Suzuki
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (T.S.); (Y.H.)
| | - Yuichi Hiraoka
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (T.S.); (Y.H.)
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Tomoko Kaneko-Ishino
- Faculty of Nursing, School of Medicine, Tokai University, Isehara 259-1193, Japan
| |
Collapse
|
13
|
Shiura H, Kitazawa M, Ishino F, Kaneko-Ishino T. Roles of retrovirus-derived PEG10 and PEG11/RTL1 in mammalian development and evolution and their involvement in human disease. Front Cell Dev Biol 2023; 11:1273638. [PMID: 37842090 PMCID: PMC10570562 DOI: 10.3389/fcell.2023.1273638] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023] Open
Abstract
PEG10 and PEG11/RTL1 are paternally expressed, imprinted genes that play essential roles in the current eutherian developmental system and are therefore associated with developmental abnormalities caused by aberrant genomic imprinting. They are also presumed to be retrovirus-derived genes with homology to the sushi-ichi retrotransposon GAG and POL, further expanding our comprehension of mammalian evolution via the domestication (exaptation) of retrovirus-derived acquired genes. In this manuscript, we review the importance of PEG10 and PEG11/RTL1 in genomic imprinting research via their functional roles in development and human disease, including neurodevelopmental disorders of genomic imprinting, Angelman, Kagami-Ogata and Temple syndromes, and the impact of newly inserted DNA on the emergence of newly imprinted regions. We also discuss their possible roles as ancestors of other retrovirus-derived RTL/SIRH genes that likewise play important roles in the current mammalian developmental system, such as in the placenta, brain and innate immune system.
Collapse
Affiliation(s)
- Hirosuke Shiura
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi, Japan
| | - Moe Kitazawa
- School of BioSciences, Faculty of Science, The University of Melbourne, Melbourne, VIC, Australia
| | - Fumitoshi Ishino
- Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tomoko Kaneko-Ishino
- Faculty of Nursing, School of Medicine, Tokai University, Isehara, Kanagawa, Japan
| |
Collapse
|
14
|
Kaneko-Ishino T, Ishino F. Retrovirus-Derived RTL/SIRH: Their Diverse Roles in the Current Eutherian Developmental System and Contribution to Eutherian Evolution. Biomolecules 2023; 13:1436. [PMID: 37892118 PMCID: PMC10604271 DOI: 10.3390/biom13101436] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
Eutherians have 11 retrotransposon Gag-like (RTL)/sushi-ichi retrotransposon homolog (SIRH) genes presumably derived from a certain retrovirus. Accumulating evidence indicates that the RTL/SIRH genes play a variety of roles in the current mammalian developmental system, such as in the placenta, brain, and innate immune system, in a eutherian-specific manner. It has been shown that the functional role of Paternally Expressed 10 (PEG10) in placental formation is unique to the therian mammals, as are the eutherian-specific roles of PEG10 and PEG11/RTL1 in maintaining the fetal capillary network and the endocrine regulation of RTL7/SIRH7 (aka Leucine Zipper Down-Regulated in Cancer 1 (LDOCK1)) in the placenta. In the brain, PEG11/RTL1 is expressed in the corticospinal tract and hippocampal commissure, mammalian-specific structures, and in the corpus callosum, a eutherian-specific structure. Unexpectedly, at least three RTL/SIRH genes, RTL5/SIRH8, RTL6/SIRH3, and RTL9/SIRH10, play important roles in combating a variety of pathogens, namely viruses, bacteria, and fungi, respectively, suggesting that the innate immunity system of the brain in eutherians has been enhanced by the emergence of these new components. In this review, we will summarize the function of 10 out of the 11 RTL/SIRH genes and discuss their roles in eutherian development and evolution.
Collapse
Affiliation(s)
- Tomoko Kaneko-Ishino
- Faculty of Nursing, School of Medicine, Tokai University, Kanagawa 259-1193, Japan
| | - Fumitoshi Ishino
- Center for Experimental Animals, Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| |
Collapse
|
15
|
Henriques WS, Young JM, Nemudryi A, Nemudraia A, Wiedenheft B, Malik HS. The diverse evolutionary histories of domesticated metaviral capsid genes in mammals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.17.558119. [PMID: 37745568 PMCID: PMC10516033 DOI: 10.1101/2023.09.17.558119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Selfish genetic elements and their remnants comprise at least half of the human genome. Active transposons duplicate by inserting copies at new sites in a host genome. Following insertion, transposons can acquire mutations that render them inactive; the accrual of additional mutations can render them unrecognizable over time. However, in rare instances, segments of transposons become useful for the host, in a process called gene domestication. Using the first complete human genome assembly and 25 additional vertebrate genomes, we analyzed the evolutionary trajectories and functional potential of genes domesticated from the capsid genes of Metaviridae, a retroviral-like retrotransposon family. Our analysis reveals four families of domesticated capsid genes in placental mammals with varied evolutionary outcomes, ranging from universal retention to lineage-specific duplications or losses and from purifying selection to lineage-specific rapid evolution. The four families of domesticated capsid genes have divergent amino-terminal domains, inherited from four distinct ancestral metaviruses. Structural predictions reveal that many domesticated genes encode a previously unrecognized RNA-binding domain retained in multiple paralogs in mammalian genomes both adjacent to and independent from the capsid domain. Collectively, our study reveals diverse outcomes of domestication of diverse metaviruses, which led to structurally and evolutionarily diverse genes that encode important, but still largely-unknown functions in placental mammals. (207).
Collapse
Affiliation(s)
- William S. Henriques
- Department of Microbiology and Cell Biology, Montana State University, Bozeman MT 59717
| | - Janet M. Young
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
| | - Artem Nemudryi
- Department of Microbiology and Cell Biology, Montana State University, Bozeman MT 59717
| | - Anna Nemudraia
- Department of Microbiology and Cell Biology, Montana State University, Bozeman MT 59717
| | - Blake Wiedenheft
- Department of Microbiology and Cell Biology, Montana State University, Bozeman MT 59717
| | - Harmit S. Malik
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
- Howard Hughes Medical Institute, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
| |
Collapse
|
16
|
Divyajanani S, Harithpriya K, Ganesan K, Ramkumar KM. Dietary Polyphenols Remodel DNA Methylation Patterns of NRF2 in Chronic Disease. Nutrients 2023; 15:3347. [PMID: 37571283 PMCID: PMC10420661 DOI: 10.3390/nu15153347] [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: 06/15/2023] [Revised: 07/17/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
The nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor crucial in regulating cellular homeostasis and apoptosis. The NRF2 gene has been implicated in various biological activities, including antioxidant, anti-inflammatory, and anticancer properties. NRF2 can be regulated genetically and epigenetically at the transcriptional, post-transcriptional, and translational levels. Although DNA methylation is one of the critical biological processes vital for gene expression, sometimes, anomalous methylation patterns result in the dysregulation of genes and consequent diseases and disorders. Several studies have reported promoter hypermethylation downregulated NRF2 expression and its downstream targets. In contrast to the unalterable nature of genetic patterns, epigenetic changes can be reversed, opening up new possibilities in developing therapies for various metabolic disorders and diseases. This review discusses the current state of the NRF2-mediated antioxidative and chemopreventive activities of several natural phytochemicals, including sulforaphane, resveratrol, curcumin, luteolin, corosolic acid, apigenin, and most other compounds that have been found to activate NRF2. This epigenetic reversal of hypermethylated NRF2 states provides new opportunities for research into dietary phytochemistry that affects the human epigenome and the possibility for cutting-edge approaches to target NRF2-mediated signaling to prevent chronic disorders.
Collapse
Affiliation(s)
- Srinivasaragavan Divyajanani
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603210, Tamil Nadu, India; (S.D.); (K.H.)
| | - Kannan Harithpriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603210, Tamil Nadu, India; (S.D.); (K.H.)
| | - Kumar Ganesan
- School of Chinese Medicine, LKS Faculty of Medicine, University of Hong Kong, 3 Sassoon Road, Hong Kong, China;
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603210, Tamil Nadu, India; (S.D.); (K.H.)
| |
Collapse
|
17
|
Richard Albert J, Kobayashi T, Inoue A, Monteagudo-Sánchez A, Kumamoto S, Takashima T, Miura A, Oikawa M, Miura F, Takada S, Hirabayashi M, Korthauer K, Kurimoto K, Greenberg MVC, Lorincz M, Kobayashi H. Conservation and divergence of canonical and non-canonical imprinting in murids. Genome Biol 2023; 24:48. [PMID: 36918927 PMCID: PMC10012579 DOI: 10.1186/s13059-023-02869-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 02/09/2023] [Indexed: 03/15/2023] Open
Abstract
BACKGROUND Genomic imprinting affects gene expression in a parent-of-origin manner and has a profound impact on complex traits including growth and behavior. While the rat is widely used to model human pathophysiology, few imprinted genes have been identified in this murid. To systematically identify imprinted genes and genomic imprints in the rat, we use low input methods for genome-wide analyses of gene expression and DNA methylation to profile embryonic and extraembryonic tissues at allele-specific resolution. RESULTS We identify 14 and 26 imprinted genes in these tissues, respectively, with 10 of these genes imprinted in both tissues. Comparative analyses with mouse reveal that orthologous imprinted gene expression and associated canonical DNA methylation imprints are conserved in the embryo proper of the Muridae family. However, only 3 paternally expressed imprinted genes are conserved in the extraembryonic tissue of murids, all of which are associated with non-canonical H3K27me3 imprints. The discovery of 8 novel non-canonical imprinted genes unique to the rat is consistent with more rapid evolution of extraembryonic imprinting. Meta-analysis of novel imprinted genes reveals multiple mechanisms by which species-specific imprinted expression may be established, including H3K27me3 deposition in the oocyte, the appearance of ZFP57 binding motifs, and the insertion of endogenous retroviral promoters. CONCLUSIONS In summary, we provide an expanded list of imprinted loci in the rat, reveal the extent of conservation of imprinted gene expression, and identify potential mechanisms responsible for the evolution of species-specific imprinting.
Collapse
Affiliation(s)
| | - Toshihiro Kobayashi
- Division of Mammalian Embryology, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Azusa Inoue
- YCI Laboratory for Metabolic Epigenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | | | - Soichiro Kumamoto
- NODAI Genome Research Center, Tokyo University of Agriculture, Tokyo, Japan
| | | | - Asuka Miura
- NODAI Genome Research Center, Tokyo University of Agriculture, Tokyo, Japan
| | - Mami Oikawa
- Division of Mammalian Embryology, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Fumihito Miura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Shuji Takada
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Masumi Hirabayashi
- Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Japan
| | - Keegan Korthauer
- Department of Statistics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kazuki Kurimoto
- Department of Embryology, Nara Medical University, Nara, Japan
| | | | - Matthew Lorincz
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
18
|
Cao W, Douglas KC, Samollow PB, VandeBerg JL, Wang X, Clark AG. Origin and Evolution of Marsupial-specific Imprinting Clusters Through Lineage-specific Gene Duplications and Acquisition of Promoter Differential Methylation. Mol Biol Evol 2023; 40:msad022. [PMID: 36721950 PMCID: PMC9937046 DOI: 10.1093/molbev/msad022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/08/2023] [Accepted: 01/25/2023] [Indexed: 02/02/2023] Open
Abstract
Genomic imprinting is a parent-of-origin-specific expression phenomenon that plays fundamental roles in many biological processes. In animals, imprinting is only observed in therian mammals, with ∼200 imprinted genes known in humans and mice. The imprinting pattern in marsupials has been minimally investigated by examining orthologs to known eutherian imprinted genes. To identify marsupial-specific imprinting in an unbiased way, we performed RNA-seq studies on samples of fetal brain and placenta from the reciprocal cross progeny of two laboratory opossum stocks. We inferred allele-specific expression for >3,000 expressed genes and discovered/validated 13 imprinted genes, including three previously known imprinted genes, Igf2r, Peg10, and H19. We estimate that marsupials imprint ∼60 autosomal genes, which is a much smaller set compared with eutherians. Among the nine novel imprinted genes, three noncoding RNAs have no known homologs in eutherian mammals, while the remaining genes have important functions in pluripotency, transcription regulation, nucleolar homeostasis, and neural differentiation. Methylation analyses at promoter CpG islands revealed differentially methylated regions in five of these marsupial-specific imprinted genes, suggesting that differential methylation is a common mechanism in the epigenetic regulation of marsupial imprinting. Clustering and co-regulation were observed at marsupial imprinting loci Pou5f3-Npdc1 and Nkrfl-Ipncr2, but eutherian-type multi-gene imprinting clusters were not detected. Also differing from eutherian mammals, the brain and placenta imprinting profiles are remarkably similar in opossums, presumably due to the shared origin of these organs from the trophectoderm. Our results contribute to a fuller understanding of the origin, evolution, and mechanisms of genomic imprinting in therian mammals.
Collapse
Affiliation(s)
- Wenqi Cao
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
- Innovation, and Commerce, Alabama Agricultural Experiment Station, Auburn University Center for Advanced Science, Auburn, AL, USA
| | - Kory C Douglas
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Paul B Samollow
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - John L VandeBerg
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Xu Wang
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
- Innovation, and Commerce, Alabama Agricultural Experiment Station, Auburn University Center for Advanced Science, Auburn, AL, USA
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Andrew G Clark
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| |
Collapse
|
19
|
Li T, Yin L, Stoll CE, Lisch D, Zhao M. Conserved noncoding sequences and de novo Mutator insertion alleles are imprinted in maize. PLANT PHYSIOLOGY 2023; 191:299-316. [PMID: 36173333 PMCID: PMC9806621 DOI: 10.1093/plphys/kiac459] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 08/30/2022] [Indexed: 05/20/2023]
Abstract
Genomic imprinting is an epigenetic phenomenon in which differential allele expression occurs in a parent-of-origin-dependent manner. Imprinting in plants is tightly linked to transposable elements (TEs), and it has been hypothesized that genomic imprinting may be a consequence of demethylation of TEs. Here, we performed high-throughput sequencing of ribonucleic acids from four maize (Zea mays) endosperms that segregated newly silenced Mutator (Mu) transposons and identified 110 paternally expressed imprinted genes (PEGs) and 139 maternally expressed imprinted genes (MEGs). Additionally, two potentially novel paternally suppressed MEGs are associated with de novo Mu insertions. In addition, we find evidence for parent-of-origin effects on expression of 407 conserved noncoding sequences (CNSs) in maize endosperm. The imprinted CNSs are largely localized within genic regions and near genes, but the imprinting status of the CNSs are largely independent of their associated genes. Both imprinted CNSs and PEGs have been subject to relaxed selection. However, our data suggest that although MEGs were already subject to a higher mutation rate prior to their being imprinted, imprinting may be the cause of the relaxed selection of PEGs. In addition, although DNA methylation is lower in the maternal alleles of both the maternally and paternally expressed CNSs (mat and pat CNSs), the difference between the two alleles in H3K27me3 levels was only observed in pat CNSs. Together, our findings point to the importance of both transposons and CNSs in genomic imprinting in maize.
Collapse
Affiliation(s)
- Tong Li
- Department of Biology, Miami University, Oxford, Ohio 45056, USA
- State Key Laboratory of Plant Physiology and Biochemistry, National Maize Improvement Center, Department of Plant Genetics and Breeding, China Agricultural University, Beijing 100193, P.R. China
| | - Liangwei Yin
- Department of Biology, Miami University, Oxford, Ohio 45056, USA
| | - Claire E Stoll
- Department of Biology, Miami University, Oxford, Ohio 45056, USA
| | - Damon Lisch
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, Indiana 47907, USA
| | - Meixia Zhao
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, Florida 32611, USA
| |
Collapse
|
20
|
Kumar A, Emdad L, Fisher PB, Das SK. Targeting epigenetic regulation for cancer therapy using small molecule inhibitors. Adv Cancer Res 2023; 158:73-161. [PMID: 36990539 DOI: 10.1016/bs.acr.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Cancer cells display pervasive changes in DNA methylation, disrupted patterns of histone posttranslational modification, chromatin composition or organization and regulatory element activities that alter normal programs of gene expression. It is becoming increasingly clear that disturbances in the epigenome are hallmarks of cancer, which are targetable and represent attractive starting points for drug creation. Remarkable progress has been made in the past decades in discovering and developing epigenetic-based small molecule inhibitors. Recently, epigenetic-targeted agents in hematologic malignancies and solid tumors have been identified and these agents are either in current clinical trials or approved for treatment. However, epigenetic drug applications face many challenges, including low selectivity, poor bioavailability, instability and acquired drug resistance. New multidisciplinary approaches are being designed to overcome these limitations, e.g., applications of machine learning, drug repurposing, high throughput virtual screening technologies, to identify selective compounds with improved stability and better bioavailability. We provide an overview of the key proteins that mediate epigenetic regulation that encompass histone and DNA modifications and discuss effector proteins that affect the organization of chromatin structure and function as well as presently available inhibitors as potential drugs. Current anticancer small-molecule inhibitors targeting epigenetic modified enzymes that have been approved by therapeutic regulatory authorities across the world are highlighted. Many of these are in different stages of clinical evaluation. We also assess emerging strategies for combinatorial approaches of epigenetic drugs with immunotherapy, standard chemotherapy or other classes of agents and advances in the design of novel epigenetic therapies.
Collapse
|
21
|
Ding Y, Zou LH, Wu J, Ramakrishnan M, Gao Y, Zhao L, Zhou M. The pattern of DNA methylation alteration, and its association with the expression changes of non-coding RNAs and mRNAs in Moso bamboo under abiotic stress. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2022; 325:111451. [PMID: 36075278 DOI: 10.1016/j.plantsci.2022.111451] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/01/2022] [Accepted: 08/31/2022] [Indexed: 06/15/2023]
Abstract
Epigenetic changes play an important role in plant growth and development and in stress response. However, DNA methylation pattern and its relationship with the expression changes of non-coding RNAs and mRNAs of Moso bamboo in response to abiotic stress is still largely unknown. In this work, we used whole-genome bisulfite sequencing in combination with whole-transcriptome sequencing to analyze the DNA methylation and transcription patterns of mRNAs and non-coding RNAs in Moso bamboo under abiotic stresses such as cold, heat, ultraviolet (UV) and salinity. We found that CHH methylation in the promoter region was positively correlated with gene expression, while CHG and CHH methylations in the gene body regions were negatively associated with gene expression. Moreover, CG and CHG methylations in the promoter regions were negatively correlated with the transcript abundance of long non-coding RNAs (lncRNAs), microRNAs (miRNAs) and circular RNAs (circRNAs). Similarly, the methylation levels of three contexts in the genic regions were negatively correlated with the transcript abundance of lncRNAs and miRNAs but positively correlated with that of circRNAs. In addition, we suggested that the reduction of 21-nt and 24-nt small interfering RNA (siRNA) expression tended to increase methylation levels in the genic regions. We found that stress-responsive genes such as CRPK1, HSFB2A and CIPK were differentially methylated and expressed. Our results also proposed that DNA methylation may regulate the expression of the transcription factors (TFs) and plant hormone signalling genes such as IAA9, MYC2 and ERF110 in response to abiotic stress. This study firstly reports the abiotic stress-responsive DNA methylation pattern and its involvement of expression of coding RNAs and non-coding RNAs in Moso bamboo. The results expand the knowledge of epigenetic mechanisms in Moso bamboo under abiotic stress and support in-depth deciphering of the function of specific non-coding RNAs in future studies.
Collapse
Affiliation(s)
- Yiqian Ding
- The State Key Laboratory of Subtropical Silviculture; Institute of Bamboo, Zhejiang A&F University, Lin'an, Hangzhou 311300, China
| | - Long-Hai Zou
- The State Key Laboratory of Subtropical Silviculture; Institute of Bamboo, Zhejiang A&F University, Lin'an, Hangzhou 311300, China.
| | - Jiajun Wu
- The State Key Laboratory of Subtropical Silviculture; Institute of Bamboo, Zhejiang A&F University, Lin'an, Hangzhou 311300, China
| | - Muthusamy Ramakrishnan
- The State Key Laboratory of Subtropical Silviculture; Institute of Bamboo, Zhejiang A&F University, Lin'an, Hangzhou 311300, China
| | - Yubang Gao
- The State Key Laboratory of Subtropical Silviculture; Institute of Bamboo, Zhejiang A&F University, Lin'an, Hangzhou 311300, China
| | - Liangzhen Zhao
- The State Key Laboratory of Subtropical Silviculture; Institute of Bamboo, Zhejiang A&F University, Lin'an, Hangzhou 311300, China; Basic Forestry and Proteomics Research Center, College of Forestry, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Mingbing Zhou
- The State Key Laboratory of Subtropical Silviculture; Institute of Bamboo, Zhejiang A&F University, Lin'an, Hangzhou 311300, China.
| |
Collapse
|
22
|
Lu X, Yang S, Jie M, Wang S, Sun C, Wu L, Chang S, Pei P, Wang S, Zhang T, Wang L. Folate deficiency disturbs PEG10 methylation modifications in human spina bifida. Pediatr Res 2022; 92:987-994. [PMID: 34934172 DOI: 10.1038/s41390-021-01908-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/26/2021] [Accepted: 12/02/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Paternally expressed gene 10 (PEG10) is believed to be a key imprinted gene involved in placenta formation. However, its role in human folate-related spina bifida (SB) remains unclear. METHODS The methylation status of the germline differentially methylated region (gDMR) in the PEG10/sarcoglycan epsilon (SGCE) imprinted cluster was compared between SB patients and control samples. Moreover, the influence of ectopic PEG10 expression on apoptosis was assessed to explore the underlying mechanisms related to folate deficiency-induced aberrant gDMR methylation in SB. RESULTS The case group exhibited a significant increase in the methylation level of the gDMR and a marked reduction in the mRNA and protein expression of PEG10 compared with the control group. A prominent negative correlation was found between the folate level in brain tissue and gDMR methylation status (r = -0.62, P = 0.001). A cell model treated with a demethylating agent showed a significant elevation of PEG10 transcription level, as well as other imprinted genes in this cluster. In addition, the inhibition of PEG10 was found to be accompanied by aberrant activation of apoptosis in SB. CONCLUSIONS Our findings suggest that disturbed gDMR methylation of the PEG10/SGCE cluster due to folate deficiency is involved in SB through aberrant activation of apoptosis. IMPACT Disturbed genomic imprinting has been verified to be involved in neural tube defects (NTDs). However, little is known about the effect of ectopic expression of imprinted gene PEG10 on human NTDs. Aberrant methylation status of the germline differentially methylated region (gDMR) of PEG10/SGCE cluster due to folate deficiency has been found to result in the inhibition of PEG10 and has a marked association with an increased occurrence of spina bifida. Inhibited expression of PEG10 partly is found to be related to the abnormal activation of apoptosis in spina bifida.
Collapse
Affiliation(s)
- Xiaolin Lu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China.,Institute of Basic Medicine, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, 100005, Beijing, China
| | - Shuyan Yang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Min Jie
- Department of Medical Genetics, Capital Institute of Pediatrics, 100020, Beijing, China
| | - Shan Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Chunrong Sun
- Laboratory of Institute, Capital Institute of Pediatrics, Beijing, China
| | - Lihua Wu
- Department of Medical Genetics, Capital Institute of Pediatrics, 100020, Beijing, China
| | - Shaoyan Chang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Pei Pei
- Laboratory of Institute, Capital Institute of Pediatrics, Beijing, China
| | - Shuowen Wang
- Institute of Basic Medicine, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, 100005, Beijing, China
| | - Ting Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Li Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China. .,Institute of Basic Medicine, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, 100005, Beijing, China.
| |
Collapse
|
23
|
Imakawa K, Kusama K, Kaneko-Ishino T, Nakagawa S, Kitao K, Miyazawa T, Ishino F. Endogenous Retroviruses and Placental Evolution, Development, and Diversity. Cells 2022; 11:cells11152458. [PMID: 35954303 PMCID: PMC9367772 DOI: 10.3390/cells11152458] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
The main roles of placentas include physical protection, nutrient and oxygen import, export of gasses and fetal waste products, and endocrinological regulation. In addition to physical protection of the fetus, the placentas must provide immune protection throughout gestation. These basic functions are well-conserved; however, placentas are undoubtedly recent evolving organs with structural and cellular diversities. These differences have been explained for the last two decades through co-opting genes and gene control elements derived from transposable elements, including endogenous retroviruses (ERVs). However, the differences in placental structures have not been explained or characterized. This manuscript addresses the sorting of ERVs and their integration into the mammalian genomes and provides new ways to explain why placental structures have diverged.
Collapse
Affiliation(s)
- Kazuhiko Imakawa
- Research Institute of Agriculture, Tokai University, Kumamoto 862-8652, Japan
- Correspondence: ; Tel.: +81-96-386-2652
| | - Kazuya Kusama
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | | | - So Nakagawa
- Department of Molecular Life Science, Tokai University School of Medicine, Nakagawa 259-1193, Japan
| | - Koichi Kitao
- Laboratory of Virus-Host Coevolution, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Takayuki Miyazawa
- Laboratory of Virus-Host Coevolution, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Fumitoshi Ishino
- Institute of Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| |
Collapse
|
24
|
Ishihara T, Hickford D, Fenelon JC, Griffith OW, Suzuki S, Renfree MB. Evolution of the short form of DNMT3A, DNMT3A2, occurred in the common ancestor of mammals. Genome Biol Evol 2022; 14:6615359. [PMID: 35749276 PMCID: PMC9254654 DOI: 10.1093/gbe/evac094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Genomic imprinting is found in marsupial and eutherian mammals, but not in monotremes. While the primary regulator of genomic imprinting in eutherians is differential DNA methylation between parental alleles, conserved imprinted genes in marsupials tend to lack DNA methylation at their promoters. DNA methylation at eutherian imprinted genes is mainly catalysed by a DNA methyltransferase (DNMT) enzyme, DNMT3A. There are two isoforms of eutherian DNMT3A: DNMT3A and DNMT3A2. DNMT3A2 is the primary isoform for establishing DNA methylation at eutherian imprinted genes and is essential for eutherian genomic imprinting. In this study, we investigated whether DNMT3A2 is also present in the two other mammalian lineages, marsupials and monotremes. We identified DNMT3A2 in both marsupials and monotremes, although imprinting has not been identified in monotremes. By analysing genomic sequences and transcriptome data across vertebrates, we concluded that the evolution of DNMT3A2 occurred in the common ancestor of mammals. In addition, DNMT3A/3A2 gene and protein expression during gametogenesis showed distinct sexual dimorphisms in a marsupial, the tammar wallaby, and this pattern coincided with the sex-specific DNA methylation reprogramming in this species as it does in mice. Our results show that DNMT3A2 is present in all mammalian groups and suggests that the basic DNMT3A/3A2-based DNA methylation mechanism is conserved at least in therian mammals.
Collapse
Affiliation(s)
- Teruhito Ishihara
- School of BioSciences, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Danielle Hickford
- School of BioSciences, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Jane C Fenelon
- School of BioSciences, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Oliver W Griffith
- Department of Biological Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Shunsuke Suzuki
- Department of Agricultural and Life Sciences, Shinshu University, Nagano, Japan
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
25
|
Renfree MB, Shaw G. Placentation in Marsupials. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2022; 234:41-60. [PMID: 34694477 DOI: 10.1007/978-3-030-77360-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
It is sometimes implied that marsupials are "aplacental," on the presumption that the only mammals that have a placenta are the eponymous "placental" mammals. This misconception has persisted despite the interest in and descriptions of the marsupial placenta, even in Amoroso's definitive chapter. It was also said that marsupials had no maternal recognition of pregnancy and no placental hormone production. In addition, it was thought that genomic imprinting could not exist in marsupials because pregnancy was so short. We now know that none of these ideas have held true with extensive studies over the last four decades definitively showing that they are indeed mammals with a fully functional placenta, and with their own specializations.
Collapse
Affiliation(s)
- Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia.
| | - Geoff Shaw
- School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
26
|
Kaneko-Ishino T, Ishino F. The Evolutionary Advantage in Mammals of the Complementary Monoallelic Expression Mechanism of Genomic Imprinting and Its Emergence From a Defense Against the Insertion Into the Host Genome. Front Genet 2022; 13:832983. [PMID: 35309133 PMCID: PMC8928582 DOI: 10.3389/fgene.2022.832983] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/11/2022] [Indexed: 12/30/2022] Open
Abstract
In viviparous mammals, genomic imprinting regulates parent-of-origin-specific monoallelic expression of paternally and maternally expressed imprinted genes (PEGs and MEGs) in a region-specific manner. It plays an essential role in mammalian development: aberrant imprinting regulation causes a variety of developmental defects, including fetal, neonatal, and postnatal lethality as well as growth abnormalities. Mechanistically, PEGs and MEGs are reciprocally regulated by DNA methylation of germ-line differentially methylated regions (gDMRs), thereby exhibiting eliciting complementary expression from parental genomes. The fact that most gDMR sequences are derived from insertion events provides strong support for the claim that genomic imprinting emerged as a host defense mechanism against the insertion in the genome. Recent studies on the molecular mechanisms concerning how the DNA methylation marks on the gDMRs are established in gametes and maintained in the pre- and postimplantation periods have further revealed the close relationship between genomic imprinting and invading DNA, such as retroviruses and LTR retrotransposons. In the presence of gDMRs, the monoallelic expression of PEGs and MEGs confers an apparent advantage by the functional compensation that takes place between the two parental genomes. Thus, it is likely that genomic imprinting is a consequence of an evolutionary trade-off for improved survival. In addition, novel genes were introduced into the mammalian genome via this same surprising and complex process as imprinted genes, such as the genes acquired from retroviruses as well as those that were duplicated by retropositioning. Importantly, these genes play essential/important roles in the current eutherian developmental system, such as that in the placenta and/or brain. Thus, genomic imprinting has played a critically important role in the evolutionary emergence of mammals, not only by providing a means to escape from the adverse effects of invading DNA with sequences corresponding to the gDMRs, but also by the acquisition of novel functions in development, growth and behavior via the mechanism of complementary monoallelic expression.
Collapse
Affiliation(s)
- Tomoko Kaneko-Ishino
- School of Medicine, Tokai University, Isehara, Japan
- *Correspondence: Tomoko Kaneko-Ishino, ; Fumitoshi Ishino,
| | - Fumitoshi Ishino
- Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
- *Correspondence: Tomoko Kaneko-Ishino, ; Fumitoshi Ishino,
| |
Collapse
|
27
|
Lestari B, Naito S, Endo A, Nishihara H, Kato A, Watanabe E, Denda K, Komada M, Fukushima T. Placental mammals acquired functional sequences in NRK for regulating the CK2-PTEN-AKT pathway and placental cell proliferation. Mol Biol Evol 2022; 39:6499274. [PMID: 34999820 PMCID: PMC8857918 DOI: 10.1093/molbev/msab371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The molecular evolution processes underlying the acquisition of the placenta in eutherian ancestors are not fully understood. Mouse NCK-interacting kinase (NIK)-related kinase (NRK) is expressed highly in the placenta and plays a role in preventing placental hyperplasia. Here, we show the molecular evolution of NRK, which confers its function for inhibiting placental cell proliferation. Comparative genome analysis identified NRK orthologs across vertebrates, which share the kinase and citron homology (CNH) domains. Evolutionary analysis revealed that NRK underwent extensive amino acid substitutions in the ancestor of placental mammals and has been since conserved. Biochemical analysis of mouse NRK revealed that the CNH domain binds to phospholipids, and a region in NRK binds to and inhibits casein kinase-2 (CK2), which we named the CK2-inhibitory region (CIR). Cell culture experiments suggest the following: 1) Mouse NRK is localized at the plasma membrane via the CNH domain, where the CIR inhibits CK2. 2) This mitigates CK2-dependent phosphorylation and inhibition of PTEN and 3) leads to the inhibition of AKT signaling and cell proliferation. Nrk deficiency increased phosphorylation levels of PTEN and AKT in mouse placenta, supporting our hypothesis. Unlike mouse NRK, chicken NRK did not bind to phospholipids and CK2, decrease phosphorylation of AKT, or inhibit cell proliferation. Both the CNH domain and CIR have evolved under purifying selection in placental mammals. Taken together, our study suggests that placental mammals acquired the phospholipid-binding CNH domain and CIR in NRK for regulating the CK2–PTEN–AKT pathway and placental cell proliferation.
Collapse
Affiliation(s)
- Beni Lestari
- School of Life Science and Technology, Tokyo Institute of Technology, Japan
| | - Satomi Naito
- School of Life Science and Technology, Tokyo Institute of Technology, Japan
| | - Akinori Endo
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Japan
| | - Hidenori Nishihara
- School of Life Science and Technology, Tokyo Institute of Technology, Japan
| | - Akira Kato
- School of Life Science and Technology, Tokyo Institute of Technology, Japan
| | - Erika Watanabe
- School of Life Science and Technology, Tokyo Institute of Technology, Japan
| | - Kimitoshi Denda
- School of Life Science and Technology, Tokyo Institute of Technology, Japan
| | - Masayuki Komada
- School of Life Science and Technology, Tokyo Institute of Technology, Japan.,Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Japan
| | - Toshiaki Fukushima
- School of Life Science and Technology, Tokyo Institute of Technology, Japan.,Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Japan
| |
Collapse
|
28
|
Identifying regulators of parental imprinting by CRISPR/Cas9 screening in haploid human embryonic stem cells. Nat Commun 2021; 12:6718. [PMID: 34795250 PMCID: PMC8602306 DOI: 10.1038/s41467-021-26949-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 10/28/2021] [Indexed: 12/13/2022] Open
Abstract
In mammals, imprinted genes are regulated by differentially methylated regions (DMRs) that are inherited from germ cells, leading to monoallelic expression in accordance with parent-of-origin. Yet, it is largely unknown how imprinted DMRs are maintained in human embryos despite global DNA demethylation following fertilization. Here, we explored the mechanisms involved in imprinting regulation by employing human parthenogenetic embryonic stem cells (hpESCs), which lack paternal alleles. We show that although global loss of DNA methylation in hpESCs affects most imprinted DMRs, many paternally-expressed genes (PEGs) remain repressed. To search for factors regulating PEGs, we performed a genome-wide CRISPR/Cas9 screen in haploid hpESCs. This revealed ATF7IP as an essential repressor of a set of PEGs, which we further show is also required for silencing sperm-specific genes. Our study reinforces an important role for histone modifications in regulating imprinted genes and suggests a link between parental imprinting and germ cell identity. Genetic imprinting ensures monoallelic gene expression critical for normal embryonic development. Here the authors take advantage of human haploid parthenogenic embryonic stem cells lacking paternal alleles to identify, by genome-wide screening, factors involved in the regulation of imprinted genes.
Collapse
|
29
|
Senft AD, Macfarlan TS. Transposable elements shape the evolution of mammalian development. Nat Rev Genet 2021; 22:691-711. [PMID: 34354263 DOI: 10.1038/s41576-021-00385-1] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2021] [Indexed: 02/06/2023]
Abstract
Transposable elements (TEs) promote genetic innovation but also threaten genome stability. Despite multiple layers of host defence, TEs actively shape mammalian-specific developmental processes, particularly during pre-implantation and extra-embryonic development and at the maternal-fetal interface. Here, we review how TEs influence mammalian genomes both directly by providing the raw material for genetic change and indirectly via co-evolving TE-binding Krüppel-associated box zinc finger proteins (KRAB-ZFPs). Throughout mammalian evolution, individual activities of ancient TEs were co-opted to enable invasive placentation that characterizes live-born mammals. By contrast, the widespread activity of evolutionarily young TEs may reflect an ongoing co-evolution that continues to impact mammalian development.
Collapse
Affiliation(s)
- Anna D Senft
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, MD, USA.
| | - Todd S Macfarlan
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
30
|
Saintilnord WN, Fondufe-Mittendorf Y. Arsenic-induced epigenetic changes in cancer development. Semin Cancer Biol 2021; 76:195-205. [PMID: 33798722 PMCID: PMC8481342 DOI: 10.1016/j.semcancer.2021.03.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/29/2022]
Abstract
Arsenic is a ubiquitous metalloid whose high levels of toxicity pose major health concerns to millions of people worldwide by increasing susceptibility to various cancers and non-cancer illnesses. Since arsenic is not a mutagen, the mechanism by which it causes changes in gene expression and disease pathogenesis is not clear. One possible mechanism is through generation of reactive oxygen species. Another equally important mechanism still very much in its infancy is epigenetic dysregulation. In this review, we discuss recent discoveries underlying arsenic-induced epigenetic changes in cancer development. Importantly, we highlight the proposed mechanisms targeted by arsenic to drive oncogenic gene expression.
Collapse
Affiliation(s)
- Wesley N Saintilnord
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA.
| | | |
Collapse
|
31
|
Shiura H, Ono R, Tachibana S, Kohda T, Kaneko-Ishino T, Ishino F. PEG10 viral aspartic protease domain is essential for the maintenance of fetal capillary structure in the mouse placenta. Development 2021; 148:272286. [PMID: 34559199 PMCID: PMC8497776 DOI: 10.1242/dev.199564] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/16/2021] [Indexed: 12/13/2022]
Abstract
The therian-specific gene paternally expressed 10 (Peg10) plays an essential role in placenta formation: Peg10 knockout mice exhibit early embryonic lethality as a result of severe placental defects. The PEG10 protein exhibits homology with long terminal repeat (LTR) retrotransposon GAG and POL proteins; therefore, we generated mice harboring a mutation in the highly conserved viral aspartic protease motif in the POL-like region of PEG10 because this motif is essential for the life cycle of LTR retrotransposons/retroviruses. Intriguingly, frequent perinatal lethality, not early embryonic lethality, was observed with fetal and placental growth retardation starting mid-gestation. In the mutant placentas, severe defects were observed in the fetal vasculature, where PEG10 is expressed in the three trophoblast cell layers that surround fetal capillary endothelial cells. Thus, Peg10 has essential roles, not only in early placenta formation, but also in placental vasculature maintenance from mid- to late-gestation. This implies that along the feto-maternal placenta interface an interaction occurs between two retrovirus-derived genes, Peg10 and retrotransposon Gag like 1 (Rtl1, also called Peg11), that is essential for the maintenance of fetal capillary endothelial cells. Summary: Disruption of the highly conserved viral aspartic protease domain in PEG10 causes placental abnormality leading to perinatal lethality in mice.
Collapse
Affiliation(s)
- Hirosuke Shiura
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi 400-8510, Japan.,Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Ryuichi Ono
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences (NIHS), Kanagawa 210-9501, Japan
| | - Saori Tachibana
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Takashi Kohda
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi 400-8510, Japan.,Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Tomoko Kaneko-Ishino
- Faculty of Nursing, School of Medicine, Tokai University, Kanagawa 259-1193, Japan
| | - Fumitoshi Ishino
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| |
Collapse
|
32
|
Association of allele-specific methylation of the ASNS gene with asparaginase sensitivity and prognosis in T-ALL. Blood Adv 2021; 6:212-224. [PMID: 34535013 PMCID: PMC8753197 DOI: 10.1182/bloodadvances.2021004271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/05/2021] [Indexed: 12/02/2022] Open
Abstract
Allele-specific methylation of the ASNS gene is associated with asparaginase sensitivity and therapeutic outcome in T-ALL. Pediatric T-ALL patients with poor prognostic SPI1 fusion exclusively exhibited ASNS hypomethylation status.
Asparaginase therapy is a key component of chemotherapy for patients with T-cell acute lymphoblastic leukemia (T-ALL). Asparaginase depletes serum asparagine by deamination into aspartic acid. Normal hematopoietic cells can survive due to asparagine synthetase (ASNS) activity, whereas leukemia cells are supposed to undergo apoptosis due to silencing of the ASNS gene. Because the ASNS gene has a typical CpG island in its promoter, its methylation status in T-ALL cells may be associated with asparaginase sensitivity. Thus, we investigated the significance of ASNS methylation status in asparaginase sensitivity of T-ALL cell lines and prognosis of childhood T-ALL. Sequencing of bisulfite polymerase chain reaction products using next-generation sequencing technology in 22 T-ALL cell lines revealed a stepwise allele-specific methylation of the ASNS gene, in association with an aberrant methylation of a 7q21 imprinted gene cluster. T-ALL cell lines with ASNS hypermethylation status showed significantly higher in vitro l-asparaginase sensitivity in association with insufficient asparaginase-induced upregulation of ASNS gene expression and lower basal ASNS protein expression. A comprehensive analysis of diagnostic samples from pediatric patients with T-ALL in Japanese cohorts (N = 77) revealed that methylation of the ASNS gene was associated with an aberrant methylation of the 7q21 imprinted gene cluster. In pediatric T-ALL patients in Japanese cohorts (n = 75), ASNS hypomethylation status was significantly associated with poor therapeutic outcome, and all cases with poor prognostic SPI1 fusion exclusively exhibited ASNS hypomethylation status. These observations show that ASNS hypomethylation status is associated with asparaginase resistance and is a poor prognostic biomarker in childhood T-ALL.
Collapse
|
33
|
Liang Y, Hu L, Li J, Liu F, Jones KC, Li D, Liu J, Chen D, You J, Yu Z, Zhang G, Dong G, Ma H. Short-term personal PM 2.5 exposure and change in DNA methylation of imprinted genes: Panel study of healthy young adults in Guangzhou city, China. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 275:116601. [PMID: 33549891 DOI: 10.1016/j.envpol.2021.116601] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 05/28/2023]
Abstract
DNA methylation (DNAm) plays a significant role in deleterious health effects inflicted by fine particulate matter (PM2.5) on the human body. Recent studies have reported that DNAm of imprinted control regions (ICRs) in imprinted genes may be a sensitive biomarker of environmental exposure. Less is known about specific biomarkers of imprinted genes after PM2.5 exposure. The relationship between PM2.5 and its chemical constituents and DNAm of ICRs in imprinted genes after short-term exposure was investigated to determine specific human biomarkers of its adverse health effects. A panel study was carried out in healthy young people in Guangzhou, China. Mixed-effects models were used to evaluate the influence of PM2.5 and its constituent exposure on DNAm while controlling for potential confounders. There was no significant correlation between DNAm and personal PM2.5 exposure mass. DNAm changes in eight ICRs (L3MBTL1, NNAT, PEG10, GNAS Ex1A, MCTS2, SNURF/SNRPN, IGF2R, and RB1) and a non-imprinted gene (CYP1B1) were significantly associated with PM2.5 constituents. Compared to non-imprinted genes, imprinted gene methylation was more susceptible to interference with PM2.5 constituent exposure. Among those genes, L3MBTL1 was the most sensitive to personal PM2.5 constituent exposure. Moreover, transition metals derived from traffic sources (Cd, Fe, Mn, and Ni) significantly influenced DNAm of the imprinted genes, suggesting the importance of more targeted measures to reduce toxic constituents. Bioinformatics analysis indicated that imprinted genes (RB1) may be correlated with pathways and diseases (non-small cell lung cancer, glioma, and bladder cancer). The present study suggests that screening the imprinted gene for DNAm can be used as a sensitive biomarker of PM2.5 exposure. The results will provide data for prevention of PM2.5 exposure and a novel perspective on potential mechanisms on an epigenetic level.
Collapse
Affiliation(s)
- Yaohui Liang
- State Key Laboratory of Organic Geochemistry and Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou, 510640, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liwen Hu
- Department of Preventive Medicine, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jun Li
- State Key Laboratory of Organic Geochemistry and Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou, 510640, China
| | - Fei Liu
- School of Business Administration, South China University of Technology, Guangzhou, 510641, China
| | - Kevin C Jones
- Lancaster Environmental Centre, Lancaster University, LA1 4YQ, Lancaster, United Kingdom
| | - Daochuan Li
- Department of Preventive Medicine, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jing Liu
- Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Duohong Chen
- Guangdong Environmental Monitoring Center, State Environmental Protection Key Laboratory of Regional Air Quality Monitoring, Guangdong Environmental Protection Key Laboratory of Atmospheric Secondary Pollution, Guangzhou, 510308, China
| | - Jing You
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, China
| | - Zhiqiang Yu
- State Key Laboratory of Organic Geochemistry and Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou, 510640, China
| | - Gan Zhang
- State Key Laboratory of Organic Geochemistry and Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou, 510640, China
| | - Guanghui Dong
- Department of Preventive Medicine, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Huimin Ma
- State Key Laboratory of Organic Geochemistry and Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou, 510640, China; Lancaster Environmental Centre, Lancaster University, LA1 4YQ, Lancaster, United Kingdom.
| |
Collapse
|
34
|
Association of aberrant ASNS imprinting with asparaginase sensitivity and chromosomal abnormality in childhood BCP-ALL. Blood 2021; 136:2319-2333. [PMID: 32573712 DOI: 10.1182/blood.2019004090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 05/18/2020] [Indexed: 02/08/2023] Open
Abstract
Karyotype is an important prognostic factor in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL), but the underlying pharmacogenomics remain unknown. Asparaginase is an integral component in current chemotherapy for childhood BCP-ALL. Asparaginase therapy depletes serum asparagine. Normal hematopoietic cells can produce asparagine by asparagine synthetase (ASNS) activity, but ALL cells are unable to synthesize adequate amounts of asparagine. The ASNS gene has a typical CpG island in its promoter. Thus, methylation of the ASNS CpG island could be one of the epigenetic mechanisms for ASNS gene silencing in BCP-ALL. To gain deep insights into the pharmacogenomics of asparaginase therapy, we investigated the association of ASNS methylation status with asparaginase sensitivity. The ASNS CpG island is largely unmethylated in normal hematopoietic cells, but it is allele-specifically methylated in BCP-ALL cells. The ASNS gene is located at 7q21, an evolutionally conserved imprinted gene cluster. ASNS methylation in childhood BCP-ALL is associated with an aberrant methylation of the imprinted gene cluster at 7q21. Aberrant methylation of mouse Asns and a syntenic imprinted gene cluster is also confirmed in leukemic spleen samples from ETV6-RUNX1 knockin mice. In 3 childhood BCP-ALL cohorts, ASNS is highly methylated in BCP-ALL patients with favorable karyotypes but is mostly unmethylated in BCP-ALL patients with poor prognostic karyotypes. Higher ASNS methylation is associated with higher L-asparaginase sensitivity in BCP-ALL through lower ASNS gene and protein expression levels. These observations demonstrate that silencing of the ASNS gene as a result of aberrant imprinting is a pharmacogenetic mechanism for the leukemia-specific activity of asparaginase therapy in BCP-ALL.
Collapse
|
35
|
Zhang X, Muglia LJ. Baby's best Foe-riend: Endogenous retroviruses and the evolution of eutherian reproduction. Placenta 2021; 113:1-7. [PMID: 33685754 DOI: 10.1016/j.placenta.2021.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/07/2021] [Accepted: 02/18/2021] [Indexed: 11/18/2022]
Abstract
High maternal investment in pregnancy and the perinatal period are prominent features of eutherian reproduction. Viviparity increases offspring survival, favoring high maternal prenatal investment. Matrotrophy through the placenta reduces maternal investment at early pregnancy, allowing the mother to abort embryos of subpar quality, therefore reducing resources wastage. On the other hand, intimate maternal-fetal interplay enables the fetus to manipulate maternal physiology to acquire more resources. This parent-offspring conflict likely drives the evolution of eutherian placentation, which is facilitated by the endogenous retroviruses (ERVs), ancient retroviruses that invaded host genome millions of years ago. ERVs bring new genes and novel regulatory elements into host genome, contribute to maternal-fetal tolerance, placenta-specific cell type formation, trophoblast gene expression network rewiring, and the establishment of imprinting. However, retroviruses/ERVs can function as infectious pathogens that interfere with host immune and inflammation pathways and cause genomic instability. In addition, ERVs coopted for host function may contribute to pathogenesis during infections due to their susceptibility to mechanisms activated by the invading pathogens. ERVs have been implicated in multiple perinatal adverse outcomes, therefore, eutherians must have evolved control mechanisms to regulate their function. Here we propose the TRIM family as an important participant of host antiviral defense and a likely candidate that mediates the coevolution of ERVs and their eutherian host. TRIMs have been shown to interact with retroviruses during each step of the infectious cycle. Understanding TRIMs' role in ERV regulation in the placenta may provide insight to both the physiology and pathology of eutherian reproduction.
Collapse
Affiliation(s)
- Xuzhe Zhang
- March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati, OH, USA; Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Louis J Muglia
- March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati, OH, USA; Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Burroughs Wellcome Fund, Research Triangle Park, NC, USA.
| |
Collapse
|
36
|
The Landscape of Genomic Imprinting at the Porcine SGCE/ PEG10 Locus from Methylome and Transcriptome of Parthenogenetic Embryos. G3-GENES GENOMES GENETICS 2020; 10:4037-4047. [PMID: 32878957 PMCID: PMC7642923 DOI: 10.1534/g3.120.401425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In mammals, imprinted genes often exist in the form of clusters in specific chromosome regions. However, in pigs, genomic imprinting of a relatively few genes and clusters has been identified, and genes within or adjacent to putative imprinted clusters need to be investigated including those at the SGCE/PEG10 locus. The objective of this study was to, using porcine parthenogenetic embryos, investigate imprinting status of genes within the genomic region spans between the COL1A2 and ASB4 genes in chromosome 9. Whole-genome bisulfite sequencing (WGBS) and RNA sequencing (RNA-seq) were conducted with normal and parthenogenetic embryos, and methylome and transcriptome were analyzed. As a result, differentially methylated regions (DMRs) between the embryos were identified, and parental allele-specific expressions of the SGCE and PEG10 genes were verified. The pig imprinted interval was limited between SGCE and PEG10, since both the COL1A2 and CASD1 genes at the centromere-proximal region and the genes between PPP1R9A and ASB4 toward the telomere were non-imprinted and biallelically expressed. Consequently, our combining analyses of methylome, transcriptome, and informative polymorphisms revealed the boundary of imprinting cluster at the SGCE/PEG10 locus in pig chromosome 9 and consolidated the landscape of genomic imprinting in pigs.
Collapse
|
37
|
Edwards CA, Takahashi N, Corish JA, Ferguson-Smith AC. The origins of genomic imprinting in mammals. Reprod Fertil Dev 2020; 31:1203-1218. [PMID: 30615843 DOI: 10.1071/rd18176] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 10/01/2018] [Indexed: 12/13/2022] Open
Abstract
Genomic imprinting is a process that causes genes to be expressed according to their parental origin. Imprinting appears to have evolved gradually in two of the three mammalian subclasses, with no imprinted genes yet identified in prototheria and only six found to be imprinted in marsupials to date. By interrogating the genomes of eutherian suborders, we determine that imprinting evolved at the majority of eutherian specific genes before the eutherian radiation. Theories considering the evolution of imprinting often relate to resource allocation and recently consider maternal-offspring interactions more generally, which, in marsupials, places a greater emphasis on lactation. In eutherians, the imprint memory is retained at least in part by zinc finger protein 57 (ZFP57), a Kruppel associated box (KRAB) zinc finger protein that binds specifically to methylated imprinting control regions. Some imprints are less dependent on ZFP57invivo and it may be no coincidence that these are the imprints that are found in marsupials. Because marsupials lack ZFP57, this suggests another more ancestral protein evolved to regulate imprints in non-eutherian subclasses, and contributes to imprinting control in eutherians. Hence, understanding the mechanisms acting at imprinting control regions across mammals has the potential to provide valuable insights into our understanding of the origins and evolution of genomic imprinting.
Collapse
Affiliation(s)
- Carol A Edwards
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Nozomi Takahashi
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Jennifer A Corish
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Anne C Ferguson-Smith
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| |
Collapse
|
38
|
Kaneko-Ishino T, Ishino F. Evolution of viviparity in mammals: what genomic imprinting tells us about mammalian placental evolution. Reprod Fertil Dev 2020; 31:1219-1227. [PMID: 30625287 DOI: 10.1071/rd18127] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 10/01/2018] [Indexed: 01/12/2023] Open
Abstract
Genomic imprinting is an epigenetic mechanism of regulating parent-of-origin-specific monoallelic expression of imprinted genes in viviparous therian mammals such as eutherians and marsupials. In this review we discuss several issues concerning the relationship between mammalian viviparity and genomic imprinting, as well as the domestication of essential placental genes: why has the genomic imprinting mechanism been so widely conserved despite the evident developmental disadvantages originating from monoallelic expression? How have genomic imprinted regions been established in the course of mammalian evolution? What drove the evolution of mammalian viviparity and how have genomic imprinting and domesticated genes contributed to this process? In considering the regulatory mechanism of imprinted genes, reciprocal expression of paternally and maternally expressed genes (PEGs and MEGs respectively) and the presence of several essential imprinted genes for placental formation and maintenance, it is likely that complementary, thereby monoallelic, expression of PEGs and MEGs is an evolutionary trade-off for survival. The innovation in novel imprinted regions was associated with the emergence of imprinting control regions, suggesting that genomic imprinting arose as a genome defence mechanism against the insertion of exogenous DNA. Mammalian viviparity emerged in the period when the atmospheric oxygen concentration was the lowest (~12%) during the last 550 million years (the Phanerozoic eon), implying this low oxygen concentration was a key factor in promoting mammalian viviparity as a response to a major evolutionary pressure. Because genomic imprinting and gene domestication from retrotransposons or retroviruses are effective measures of changing genomic function in therian mammals, they are likely to play critical roles in the emergence of viviparity for longer gestation periods.
Collapse
Affiliation(s)
- Tomoko Kaneko-Ishino
- School of Health Sciences, Tokai University, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| | - Fumitoshi Ishino
- Department of Epigenetics, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| |
Collapse
|
39
|
Affiliation(s)
- Kenji Matsuura
- Laboratory of Insect Ecology, Graduate School of AgricultureKyoto University Kyoto Japan
| |
Collapse
|
40
|
Yonekura S, Ohata M, Tsuchiya M, Tokita H, Mizusawa M, Tokutake Y. Peg1/Mest, an imprinted gene, is involved in mammary gland maturation. J Cell Physiol 2019; 234:1080-1087. [PMID: 30144363 DOI: 10.1002/jcp.27219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/16/2018] [Indexed: 12/24/2022]
Abstract
Imprinted genes, which are specific to mammals, play important roles in cell proliferation, differentiation, ontogeny, and other phenomena. Moreover, these genes are considered crucial in the research of mammalian evolution. In the current study, we investigated the association between the expression of paternally imprinted gene paternally expressed 1/mesoderm-specific transcript (Peg1/Mest) and the maturation of the mammary gland. Quantitative real-time polymerase chain reaction analysis of Peg1/Mest gene expression at different stages of mouse mammary gland maturation revealed that its expression increased during gestation but decreased during lactation. Immunohistochemical staining demonstrated that Peg1/Mest was expressed in mammary epithelial cells. We measured expression levels of Peg1/Mest and E-cadherin during mammary alveoli formation using immunofluorescence staining a cell model for mammary alveoli formation in a 3D culture system. We found that the onset of E-cadherin expression roughly coincided with the peak of Peg1/Mest expression. Moreover, we discovered that the formation and proliferation of alveoli were suppressed in Peg1/Mest knockdown mammary epithelial cells. These results suggest that Peg1/Mest plays a certain role in mammary alveoli formation. To clarify the role of Peg1/Mest in the lactogenic differentiation of mammary epithelial cells, we examined the lactogenic differentiation capability of Peg1/Mest-overexpressing HC11 cells. Application of a differentiation-inducing stimulus did not increase β-casein expression in Peg1/Mest-overexpressing HC11 cells. The current study for the first time reports the involvement of an imprinted gene in mammary gland maturation.
Collapse
Affiliation(s)
- Shinichi Yonekura
- Department of Interdisciplinary Genome Sciences and Cell Metabolism, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research (ICCER), Shinshu University, Nagano, Japan
- Department of Biomedical Engineering, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
- Department of Bioscience and Food Production Science, Interdisciplinary Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Masaki Ohata
- Department of Biomedical Engineering, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Megumi Tsuchiya
- Department of Biomedical Engineering, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Hitomi Tokita
- Department of Biomedical Engineering, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Moeko Mizusawa
- Department of Biomedical Engineering, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Yukako Tokutake
- Department of Bioscience and Food Production Science, Interdisciplinary Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| |
Collapse
|
41
|
Abstract
The monoallelic parent of origin-specific expression of imprinted genes in mammals is regulated by differentially DNA methylated imprinting control regions (ICRs). In contrast to most of the genome, ICRs must maintain their DNA methylation and parental identity despite extensive epigenetic reprogramming that takes place after fertilization. Previous work demonstrated that the Krüppel-associated box (KRAB)-containing zinc finger protein (KZFP) ZFP57 protects select ICRs from demethylation and preserves parental identity. However, some loci are unaffected in Zfp57-null mice. Thus, it has been speculated that other KZFPs must be involved in this process. Takahashi and colleagues (pp. 49-54) now report one such KZFP: ZFP445.
Collapse
Affiliation(s)
- Aimee M Juan
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Marisa S Bartolomei
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
42
|
Takahashi N, Coluccio A, Thorball CW, Planet E, Shi H, Offner S, Turelli P, Imbeault M, Ferguson-Smith AC, Trono D. ZNF445 is a primary regulator of genomic imprinting. Genes Dev 2019; 33:49-54. [PMID: 30602440 PMCID: PMC6317318 DOI: 10.1101/gad.320069.118] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/07/2018] [Indexed: 12/29/2022]
Abstract
Genomic imprinting is an epigenetic process regulated by germline-derived DNA methylation, causing parental origin-specific monoallelic gene expression. Zinc finger protein 57 (ZFP57) is critical for maintenance of this epigenetic memory during post-fertilization reprogramming, yet incomplete penetrance of ZFP57 mutations in humans and mice suggests additional effectors. We reveal that ZNF445/ZFP445, which we trace to the origins of imprinting, binds imprinting control regions (ICRs) in mice and humans. In mice, ZFP445 and ZFP57 act together, maintaining all but one ICR in vivo, whereas earlier embryonic expression of ZNF445 and its intolerance to loss-of-function mutations indicate greater importance in the maintenance of human imprints.
Collapse
Affiliation(s)
- Nozomi Takahashi
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| | - Andrea Coluccio
- School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Christian W Thorball
- School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Evarist Planet
- School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Hui Shi
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| | - Sandra Offner
- School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Priscilla Turelli
- School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Michael Imbeault
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| | | | - Didier Trono
- School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne 1015, Switzerland
| |
Collapse
|
43
|
Graves JAM. Marsupial genomics meet marsupial reproduction. Reprod Fertil Dev 2018; 31:1181-1188. [PMID: 30482268 DOI: 10.1071/rd18234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/08/2018] [Indexed: 11/23/2022] Open
Abstract
We came from very different backgrounds, with different skills and interests. Marilyn Renfree was recognised as 'a giant of marsupial embryology'; I had spent my working life studying genes and chromosomes. We teamed up out of mutual respect (awe on my side) to form, with Des Cooper, the ARC Centre of Excellence in Kangaroo Genomics. This is the story of how our collaboration came to be, and what it has produced for our knowledge of some of the world's most remarkable animals.
Collapse
|
44
|
Suzuki S, Shaw G, Renfree MB. Identification of a novel antisense noncoding RNA, ALID, transcribed from the putative imprinting control region of marsupial IGF2R. Epigenetics Chromatin 2018; 11:55. [PMID: 30268152 PMCID: PMC6162910 DOI: 10.1186/s13072-018-0227-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/25/2018] [Indexed: 12/20/2022] Open
Abstract
Background Genomic imprinting leads to maternal expression of IGF2R in both mouse and opossum. In mouse, the antisense long noncoding (lnc) RNA Airn, which is paternally expressed from the differentially methylated region (DMR) in the second intron of Igf2r, is required to silence the paternal Igf2r. In opossum, however, intriguingly, the DMR was reported to be in a different downstream intron (intron 11) and there was no antisense lncRNA detected in previous analyses. Therefore, clarifying the imprinting mechanism of marsupial IGF2R is of great relevance for understanding the origin and evolution of genomic imprinting in the IGF2R locus. Thus, the antisense lncRNA associated with the marsupial DMR can be considered as the ‘missing link’. In this study, we identified a novel antisense lncRNA, ALID, after detailed analysis of the IGF2R locus in an Australian marsupial, the tammar wallaby, Macropus eugenii, and compared it to that of the grey short-tailed opossum, Monodelphis domestica. Results Tammar IGF2R showed maternal expression and had a maternally methylated CpG island (CGI) in intron 12 as well as a promoter CGI without differential methylation, but none in the second intron. Re-analysis of the IGF2R of opossum detected the CGI in intron 12, not intron 11, as previously reported, confirming that the DMR in intron 12 is conserved between these marsupials and so is the putative imprinting control region of marsupial IGF2R. ALID is paternally expressed from the middle of the DMR and is approximately 650 bp long with a single exon structure that is extremely short compared to Airn. Hence, the lncRNA transcriptional overlap of the IGF2R promoter, which is essential for the Igf2r silencing in the mouse, is likely absent in tammar. This suggests that fundamental differences in the lncRNA-based silencing mechanisms evolved in eutherian and marsupial IGF2R and may reflect the lack of differential methylation in the promoter CGI of marsupial IGF2R. Conclusions Our study thus provides the best candidate factor for establishing paternal silencing of marsupial IGF2R without transcriptional overlap, which is distinct from the Igf2r silencing mechanism of Airn, but which may be analogous to the mode of action for the flanking Slc22a2 and Slc22a3 gene silencing in the mouse placenta. Electronic supplementary material The online version of this article (10.1186/s13072-018-0227-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shunsuke Suzuki
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, 399-4598, Japan.,Department of Interdisciplinary Genome Sciences and Cell Metabolism, Institute for Biomedical Sciences, ICCER, Shinshu University, Nagano, 399-4598, Japan
| | - Geoffrey Shaw
- School of BioSciences, The University of Melbourne, Victoria, 3010, Australia
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Victoria, 3010, Australia.
| |
Collapse
|
45
|
Xie T, Pan S, Zheng H, Luo Z, Tembo KM, Jamal M, Yu Z, Yu Y, Xia J, Yin Q, Wang M, Yuan W, Zhang Q, Xiong J. PEG10 as an oncogene: expression regulatory mechanisms and role in tumor progression. Cancer Cell Int 2018; 18:112. [PMID: 30123090 PMCID: PMC6090666 DOI: 10.1186/s12935-018-0610-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 08/04/2018] [Indexed: 02/07/2023] Open
Abstract
Cancer is a major public health problem as one of the leading causes of death worldwide. Deciphering the molecular regulation mechanisms of tumor progression can make way for tumor diagnosis and therapy. Paternally expressed gene 10 (PEG10), located on human chromosome 7q21.3, has turned out to be an oncogene implicated in the proliferation, apoptosis and metastasis of tumors. PEG10 has been found to be positively expressed in a variety of cancers with seemingly complex expression regulation mechanisms. In this review, we focus on the most vital factors influencing PEG10 expression and recapitulate some of the currently known and potential mechanisms of PEG10 affecting tumor progression, as understanding the molecular regulatory mechanisms of tumor progression can provide potential PEG10 related diagnosis and biomarker specific targeted therapies.
Collapse
Affiliation(s)
- Tian Xie
- 1Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071 China
| | - Shan Pan
- 1Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071 China
| | - Hang Zheng
- 2Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Zilv Luo
- 1Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071 China
| | | | - Muhammad Jamal
- 4State Key Laboratory of Agriculture Microbiology, Huazhong Agricultural University, Wuhan, 430070 China
| | - Zhongyang Yu
- 1Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071 China
| | - Yao Yu
- 1Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071 China
| | - Jing Xia
- 1Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071 China
| | - Qian Yin
- 1Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071 China
| | - Meng Wang
- 1Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071 China
| | - Wen Yuan
- 1Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071 China
| | - Qiuping Zhang
- 1Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071 China.,5Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University, Wuhan, 430071 China
| | - Jie Xiong
- 1Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071 China
| |
Collapse
|
46
|
Liu H, Shang X, Zhu H. LncRNA/DNA binding analysis reveals losses and gains and lineage specificity of genomic imprinting in mammals. Bioinformatics 2018; 33:1431-1436. [PMID: 28052924 DOI: 10.1093/bioinformatics/btw818] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/20/2016] [Indexed: 11/14/2022] Open
Abstract
Motivation Genomic imprinting is regulated by lncRNAs and is important for embryogenesis, physiology and behaviour in mammals. Aberrant imprinting causes diseases and disorders. Experimental studies have examined genomic imprinting primarily in humans and mice, thus leaving some fundamental issues poorly addressed. The cost of experimentally examining imprinted genes in many tissues in diverse species makes computational analysis of lncRNAs' DNA binding sites valuable. Results We performed lncRNA/DNA binding analysis in imprinting clusters from multiple mammalian clades and discovered the following: (i) lncRNAs and imprinting sites show significant losses and gains and distinct lineage-specificity; (ii) binding of lncRNAs to promoters of imprinted genes may occur widely throughout the genome; (iii) a considerable number of imprinting sites occur in only evolutionarily more derived species; and (iv) multiple lncRNAs may bind to the same imprinting sites, and some lncRNAs have multiple DNA binding motifs. These results suggest that the occurrence of abundant lncRNAs in mammalian genomes makes genomic imprinting a mechanism of adaptive evolution at the epigenome level. Availability and Implementation The data and program are available at the database LongMan at lncRNA.smu.edu.cn. Contact zhuhao@smu.edu.cn. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Haihua Liu
- Bioinformatics Section, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaoxiao Shang
- Bioinformatics Section, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hao Zhu
- Bioinformatics Section, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
47
|
Taniguchi K, Kawai T, Hata K. Placental Development and Nutritional Environment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1012:63-73. [PMID: 29956195 DOI: 10.1007/978-981-10-5526-3_7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The placenta is considered to have developed recently in mammalian evolution. While the fundamental function of the placenta, i.e., providing nutrients and oxygen to the fetus and receiving waste products, is the same in all mammals, the morphology of the placenta varies substantially in a species-dependent manner. Therefore, considerable interest exists in understanding placental development and function in mammals from a molecular biological viewpoint. Numerous recent studies have shown that various environmental factors before and during pregnancy, including nutrition, affect placental formation and function and that alterations in placental formation and function can influence the developing fetus and the offspring after birth. To date, the relationship between nutrition and the placenta has been investigated in several species, various model organisms, and humans. In this chapter, we discuss the current knowledge of the placenta and the epigenome and then highlight the effects of nutrition during pregnancy on the placenta and the fetus and on the offspring after birth.
Collapse
Affiliation(s)
- Kosuke Taniguchi
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan.
| | - Tomoko Kawai
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
48
|
Zadora J, Singh M, Herse F, Przybyl L, Haase N, Golic M, Yung HW, Huppertz B, Cartwright JE, Whitley G, Johnsen GM, Levi G, Isbruch A, Schulz H, Luft FC, Müller DN, Staff AC, Hurst LD, Dechend R, Izsvák Z. Disturbed Placental Imprinting in Preeclampsia Leads to Altered Expression of DLX5, a Human-Specific Early Trophoblast Marker. Circulation 2017; 136:1824-1839. [PMID: 28904069 PMCID: PMC5671803 DOI: 10.1161/circulationaha.117.028110] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 08/28/2017] [Indexed: 01/23/2023]
Abstract
Supplemental Digital Content is available in the text. Background: Preeclampsia is a complex and common human-specific pregnancy syndrome associated with placental pathology. The human specificity provides both intellectual and methodological challenges, lacking a robust model system. Given the role of imprinted genes in human placentation and the vulnerability of imprinted genes to loss of imprinting changes, there has been extensive speculation, but no robust evidence, that imprinted genes are involved in preeclampsia. Our study aims to investigate whether disturbed imprinting contributes to preeclampsia. Methods: We first aimed to confirm that preeclampsia is a disease of the placenta by generating and analyzing genome-wide molecular data on well-characterized patient material. We performed high-throughput transcriptome analyses of multiple placenta samples from healthy controls and patients with preeclampsia. Next, we identified differentially expressed genes in preeclamptic placentas and intersected them with the list of human imprinted genes. We used bioinformatics/statistical analyses to confirm association between imprinting and preeclampsia and to predict biological processes affected in preeclampsia. Validation included epigenetic and cellular assays. In terms of human specificity, we established an in vitro invasion-differentiation trophoblast model. Our comparative phylogenetic analysis involved single-cell transcriptome data of human, macaque, and mouse preimplantation embryogenesis. Results: We found disturbed placental imprinting in preeclampsia and revealed potential candidates, including GATA3 and DLX5, with poorly explored imprinted status and no prior association with preeclampsia. As a result of loss of imprinting, DLX5 was upregulated in 69% of preeclamptic placentas. Levels of DLX5 correlated with classic preeclampsia markers. DLX5 is expressed in human but not in murine trophoblast. The DLX5high phenotype resulted in reduced proliferation, increased metabolism, and endoplasmic reticulum stress-response activation in trophoblasts in vitro. The transcriptional profile of such cells mimics the transcriptome of preeclamptic placentas. Pan-mammalian comparative analysis identified DLX5 as part of the human-specific regulatory network of trophoblast differentiation. Conclusions: Our analysis provides evidence of a true association among disturbed imprinting, gene expression, and preeclampsia. As a result of disturbed imprinting, the upregulated DLX5 affects trophoblast proliferation. Our in vitro model might fill a vital niche in preeclampsia research. Human-specific regulatory circuitry of DLX5 might help explain certain aspects of preeclampsia.
Collapse
Affiliation(s)
- Julianna Zadora
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Manvendra Singh
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Florian Herse
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Lukasz Przybyl
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Nadine Haase
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Michaela Golic
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Hong Wa Yung
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Berthold Huppertz
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Judith E Cartwright
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Guy Whitley
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Guro M Johnsen
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Giovanni Levi
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Annette Isbruch
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Herbert Schulz
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Friedrich C Luft
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Dominik N Müller
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Anne Cathrine Staff
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Laurence D Hurst
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.).
| | - Ralf Dechend
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.).
| | - Zsuzsanna Izsvák
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.).
| |
Collapse
|
49
|
De Donato M, Hussain T, Rodulfo H, Peters SO, Imumorin IG, Thomas BN. Conservation of Repeats at the Mammalian KCNQ1OT1-CDKN1C Region Suggests a Role in Genomic Imprinting. Evol Bioinform Online 2017; 13:1176934317715238. [PMID: 28659711 PMCID: PMC5476424 DOI: 10.1177/1176934317715238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/23/2017] [Indexed: 12/19/2022] Open
Abstract
KCNQ1OT1 is located in the region with the highest number of genes showing genomic imprinting, but the mechanisms controlling the genes under its influence have not been fully elucidated. Therefore, we conducted a comparative analysis of the KCNQ1/KCNQ1OT1-CDKN1C region to study its conservation across the best assembled eutherian mammalian genomes sequenced to date and analyzed potential elements that may be implicated in the control of genomic imprinting in this region. The genomic features in these regions from human, mouse, cattle, and dog show a higher number of genes and CpG islands (detected using cpgplot from EMBOSS), but lower number of repetitive elements (including short interspersed nuclear elements and long interspersed nuclear elements), compared with their whole chromosomes (detected by RepeatMasker). The KCNQ1OT1-CDKN1C region contains the highest number of conserved noncoding sequences (CNS) among mammals, where we found 16 regions containing about 38 different highly conserved repetitive elements (using mVista), such as LINE1 elements: L1M4, L1MB7, HAL1, L1M4a, L1Med, and an LTR element: MLT1H. From these elements, we found 74 CNS showing high sequence identity (>70%) between human, cattle, and mouse, from which we identified 13 motifs (using Multiple Em for Motif Elicitation/Motif Alignment and Search Tool) with a significant probability of occurrence, 3 of which were the most frequent and were used to find transcription factor-binding sites. We detected several transcription factors (using JASPAR suite) from the families SOX, FOX, and GATA. A phylogenetic analysis of these CNS from human, marmoset, mouse, rat, cattle, dog, horse, and elephant shows branches with high levels of support and very similar phylogenetic relationships among these groups, confirming previous reports. Our results suggest that functional DNA elements identified by comparative genomics in a region densely populated with imprinted mammalian genes may be related to the regulation of imprinted gene expression.
Collapse
Affiliation(s)
- Marcos De Donato
- Animal Genetics and Genomics Laboratory, Office of International Programs, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA.,Escuela de Bioingenierias, Tecnologico de Monterrey, Campus Querétaro, Santiago de Querétaro, Mexico
| | - Tanveer Hussain
- Animal Genetics and Genomics Laboratory, Office of International Programs, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA.,Department Molecular Biology, Virtual University of Pakistan, Lahore, Pakistan
| | - Hectorina Rodulfo
- Escuela de Bioingenierias, Tecnologico de Monterrey, Campus Querétaro, Santiago de Querétaro, Mexico
| | - Sunday O Peters
- Department of Animal Science, Berry College, Mount Berry, GA, USA
| | - Ikhide G Imumorin
- Animal Genetics and Genomics Laboratory, Office of International Programs, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA.,African Institute for Biosciences Research and Training, Ibadan, Nigeria.,School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Bolaji N Thomas
- Department of Biomedical Sciences, Rochester Institute of Technology, Rochester, NY, USA
| |
Collapse
|
50
|
Allen SE, Nowacki M. Necessity Is the Mother of Invention: Ciliates, Transposons, and Transgenerational Inheritance. Trends Genet 2017; 33:197-207. [PMID: 28174020 DOI: 10.1016/j.tig.2017.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 01/09/2017] [Accepted: 01/11/2017] [Indexed: 01/01/2023]
Abstract
Ciliates are a fascinating model system for the study of the interaction between eukaryotic germlines and somatic lines, especially with regard to the invasion and defence against transposable elements. They separate their germline and somatic line into two nuclei within the same cell, and they silence transposons and repetitive elements by way of deleting them from their somatic genome. This large-scale deletion event uses a series of intricate sequence targeting pathways involving small RNAs and transposases, part of which consists of a transnuclear comparison between maternal soma and daughter germline. We present recent progress in this dynamic field, and argue that these DNA targeting pathways provide an optimal system for the transgenerational inheritance of acquired traits. Ciliates thus also demonstrate the evolutionary value of transposable elements, both as sources of sequence diversity and also as drivers of adaptive evolution by necessitating defensive systems.
Collapse
Affiliation(s)
- Sarah E Allen
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Mariusz Nowacki
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland.
| |
Collapse
|