Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 1400: 95-106, 2016.
Article in English | MEDLINE | ID: mdl-26895048

ABSTRACT

With the advent of new generations of high-throughput sequencing technologies, the catalog of human genome variants created by retrotransposon activity is expanding rapidly. However, despite these advances in describing L1 diversity and the fact that L1 must retrotranspose in the germline or prior to germline partitioning to be evolutionarily successful, direct assessment of de novo L1 retrotransposition in the germline or early embryogenesis has not been achieved for endogenous L1 elements. A direct study of de novo L1 retrotransposition into susceptible loci within sperm DNA (Freeman et al., Hum Mutat 32(8):978-988, 2011) suggested that the rate of L1 retrotransposition in the germline is much lower than previously estimated (<1 in 400 individuals versus 1 in 9 individuals (Kazazian, Nat Genet 22(2):130, 1999). Based on these revised estimates of the L1 retrotransposition rate, we modified the ATLAS L1 display technique (Badge et al., Am J Hum Genet 72(4):823-838, 2003) to investigate de novo L1 retrotransposition in human genomes. In this chapter, we describe how we combined a high-coverage ATLAS variant with high-throughput sequencing, achieving 11-25× sequence depth per single amplicon, to study L1 retrotransposition in whole genome amplified (WGA) DNAs.


Subject(s)
High-Throughput Nucleotide Sequencing , Long Interspersed Nucleotide Elements , Molecular Typing , Polymerase Chain Reaction , Computational Biology/methods , Genome, Human , Genomic Library , Genomics/methods , Humans , Molecular Typing/methods , Polymerase Chain Reaction/methods
2.
Retrovirology ; 12: 35, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25927962

ABSTRACT

BACKGROUND: To date, the human population census of proviruses of the Betaretrovirus-like human endogenous retroviral (HERV-K) (HML-2) family has been compiled from a limited number of complete genomes, making it certain that rare polymorphic loci are under-represented and are yet to be described. RESULTS: Here we describe a suppression PCR-based method called genome-wide amplification of proviral sequences (GAPS) that selectively amplifies DNA fragments containing the termini of HERV-K(HML-2) proviral sequences and their flanking genomic sequences. We analysed the HERV-K(HML-2) proviral content of 101 unrelated humans, 4 common chimpanzees and three centre d'etude du polymorphisme humain (CEPH) pedigrees (44 individuals). The technique isolated HERV-K(HML-2) proviruses that had integrated in the genomes of the great apes throughout their divergence and included evolutionarily young elements still unfixed for presence/absence. CONCLUSIONS: By examining the HERV-K(HML-2) proviral content of 145 humans we detected a new insertionally polymorphic Type I HERV-K(HML-2) provirus. We also observed provirus versus solo long terminal repeat (LTR) polymorphism within humans at a previously unreported, but ancient, locus. Finally, we report two novel chimpanzee specific proviruses, one of which is dimorphic for a provirus versus solo LTR. Thus GAPS enables the isolation of uncharacterised HERV-K(HML-2) proviral sequences and provides a direct means to assess inter-individual genetic variation associated with HERV-K(HML-2) proviruses.


Subject(s)
Endogenous Retroviruses/genetics , Endogenous Retroviruses/isolation & purification , Genetic Variation , Polymerase Chain Reaction , Proviruses/genetics , Retroviridae Infections/veterinary , Retroviridae Infections/virology , Animals , DNA, Viral/chemistry , DNA, Viral/genetics , Endogenous Retroviruses/classification , Humans , Molecular Sequence Data , Pan troglodytes , Sequence Analysis, DNA
3.
Genome Biol Evol ; 6(11): 3025-38, 2014 Oct 27.
Article in English | MEDLINE | ID: mdl-25349268

ABSTRACT

ß-defensins are a family of important peptides of innate immunity, involved in host defense, immunomodulation, reproduction, and pigmentation. Genes encoding ß-defensins show evidence of birth-and-death evolution, adaptation by amino acid sequence changes, and extensive copy number variation (CNV) within humans and other species. The role of CNV in the adaptation of ß-defensins to new functions remains unclear, as does the adaptive role of CNV in general. Here, we fine-map CNV of a cluster of ß-defensins in humans and rhesus macaques. Remarkably, we found that the structure of the CNV is different between primates, with distinct mutational origins and CNV boundaries defined by retroviral long terminal repeat elements. Although the human ß-defensin CNV region is 322 kb and encompasses several genes, including ß-defensins, a long noncoding RNA gene, and testes-specific zinc-finger transcription factors, the orthologous region in the rhesus macaque shows CNV of a 20-kb region, containing only a single gene, the ortholog of the human ß-defensin-2 gene. Despite its independent origins, the range of gene copy numbers in the rhesus macaque is similar to humans. In addition, the rhesus macaque gene has been subject to divergent positive selection at the amino acid level following its initial duplication event between 3 and 9.5 Ma, suggesting adaptation of this gene as the macaque successfully colonized novel environments outside Africa. Therefore, the molecular phenotype of ß-defensin-2 CNV has undergone convergent evolution, and this gene shows evidence of adaptation at the amino acid level in rhesus macaques.


Subject(s)
Adaptation, Physiological/genetics , DNA Copy Number Variations , Evolution, Molecular , beta-Defensins/genetics , Animals , Humans , Macaca mulatta , Mutation , Selection, Genetic
4.
Hum Mutat ; 34(7): 974-85, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23553801

ABSTRACT

Long INterspersed Element-1 (LINE-1 or L1) retrotransposons are the only autonomously active transposable elements in the human genome. The average human genome contains ∼80-100 active L1s, but only a subset of these L1s are highly active or 'hot'. Human L1s are closely related in sequence, making it difficult to decipher progenitor/offspring relationships using traditional phylogenetic methods. However, L1 mRNAs can sometimes bypass their own polyadenylation signal and instead utilize fortuitous polyadenylation signals in 3' flanking genomic DNA. Retrotransposition of the resultant mRNAs then results in lineage specific sequence "tags" (i.e., 3' transductions) that mark the descendants of active L1 progenitors. Here, we developed a method (Transduction-Specific Amplification Typing of L1 Active Subfamilies or TS-ATLAS) that exploits L1 3' transductions to identify active L1 lineages in a genome-wide context. TS-ATLAS enabled the characterization of a putative active progenitor of one L1 lineage that includes the disease causing L1 insertion L1RP , and the identification of new retrotransposition events within two other "hot" L1 lineages. Intriguingly, the analysis of the newly discovered transduction lineage members suggests that L1 polyadenylation, even within a lineage, is highly stochastic. Thus, TS-ATLAS provides a new tool to explore the dynamics of L1 lineage evolution and retrotransposon biology.


Subject(s)
Genome, Human/genetics , Long Interspersed Nucleotide Elements/genetics , Mutagenesis, Insertional/methods , Retroelements/genetics , DNA/genetics , Humans , Polyadenylation
5.
Mob Genet Elements ; 1(2): 122-127, 2011 Jul.
Article in English | MEDLINE | ID: mdl-22016860

ABSTRACT

The ongoing activity of the human retrotransposon Long Interspersed Element 1 (LINE-1 or L1) continues to impact the human genome in various ways. Throughout evolution, mammalian and primate genomes have been under selection to generate strategies to reduce the activity of selfish DNA like L1. Similarly, selfish DNA has evolved to elude these containment systems. This intragenomic conflict has left many inactive versions of LINEs and other Transposable Elements (TEs) littering the human genome, which together account for roughly half of our DNA. Here, we survey the distinct mechanisms operating in the human genome that seem to reduce the mobility of L1s. In addition, we discuss recent findings that strongly suggest epigenetic mechanisms specifically regulate L1 activity in pluripotent human cells.

6.
Annu Rev Genomics Hum Genet ; 12: 187-215, 2011.
Article in English | MEDLINE | ID: mdl-21801021

ABSTRACT

The completion of the human genome reference sequence ushered in a new era for the study and discovery of human transposable elements. It now is undeniable that transposable elements, historically dismissed as junk DNA, have had an instrumental role in sculpting the structure and function of our genomes. In particular, long interspersed element-1 (LINE-1 or L1) and short interspersed elements (SINEs) continue to affect our genome, and their movement can lead to sporadic cases of disease. Here, we briefly review the types of transposable elements present in the human genome and their mechanisms of mobility. We next highlight how advances in DNA sequencing and genomic technologies have enabled the discovery of novel retrotransposons in individual genomes. Finally, we discuss how L1-mediated retrotransposition events impact human genomes.


Subject(s)
Long Interspersed Nucleotide Elements , Disease/genetics , Genetic Variation , Genome, Human , Humans , Retroelements
7.
Hum Mutat ; 32(8): 978-88, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21560187

ABSTRACT

Long interspersed nuclear element 1 (L1) retrotransposons are the only autonomously mobile human transposable elements. L1 retrotransposition has shaped our genome via insertional mutagenesis, sequence transduction, pseudogene formation, and ectopic recombination. However, L1 germline retrotransposition dynamics are poorly understood because de novo insertions occur very rarely: the frequency of disease-causing retrotransposon insertions suggests that one insertion event occurs in roughly 18-180 gametes. The method described here recovers full-length L1 insertions by using hybridization enrichment to capture L1 sequences from multiplex PCR-amplified DNA. Enrichment is achieved by hybridizing L1-specific biotinylated oligonucleotides to complementary molecules, followed by capture on streptavidin-coated paramagnetic beads. We show that multiplex, long-range PCR can amplify single molecules containing full-length L1 insertions for recovery by hybridization enrichment. We screened 600 µg of sperm DNA from one donor, but no bone fide de novo L1 insertions were found, suggesting a L1 retrotransposition frequency of <1 insertion in 400 haploid genomes. This lies below the lower bound of previous estimates, and indicates that L1 insertion, at least into the loci studied, is very rare in the male germline. It is a paradox that L1 replication is ongoing in the face of such apparently low activity.


Subject(s)
Genome, Human/genetics , Long Interspersed Nucleotide Elements/genetics , Mutagenesis, Insertional/genetics , Nucleic Acid Hybridization , DNA/genetics , Genetic Loci , Humans , Male , Polymerase Chain Reaction , Spermatozoa/metabolism
8.
Mol Biotechnol ; 47(3): 243-52, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20872285

ABSTRACT

Intracisternal A-type particle (IAP) elements are high copy number long terminal repeat (LTR) rodent retrotransposons. Some IAP elements can transpose, and are responsible for ~12% of spontaneous mouse mutations. Inbred mouse strains show variation in genomic IAP distribution, contributing to inter-strain genetic variability. Additionally IAP elements can influence the transcriptional regulation of neighbouring genes through their strong LTR promoter, effecting phenotypic variation. This genetic and phenotypic variability can translate into experimental variability between mouse strains. For example, it has been demonstrated that strain-specific genetic/epigenetic factors can interact to yield variable responses to drugs. Therefore, in experimental contexts it is essential to unequivocally identify mouse strains. Recently it was estimated that any two inbred strains share only ~40% of their IAP insertions. Of the remaining 60%, some insertions will be strain specific, fixed during inbreeding. These fixed insertions can be exploited as genetic markers to identify inbred strains, if they can be identified simply and efficiently. Here, we report the development of a PCR-based system allowing direct acquisition of strain-specific IAP insertions. In a pilot study, we identified 21 IAP loci, genotyped IAP insertions at 9 loci, and discovered two strain-specific insertions that could reliably identify these strains.


Subject(s)
Genes, Intracisternal A-Particle/genetics , Retroelements/genetics , Terminal Repeat Sequences/genetics , Animals , Genotype , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA , Polymerase Chain Reaction
9.
Mol Cell Biol ; 31(2): 300-16, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21041477

ABSTRACT

Long interspersed element 1s (LINE-1s or L1s) are a family of non-long-terminal-repeat retrotransposons that predominate in the human genome. Active LINE-1 elements encode proteins required for their mobilization. L1-encoded proteins also act in trans to mobilize short interspersed elements (SINEs), such as Alu elements. L1 and Alu insertions have been implicated in many human diseases, and their retrotransposition provides an ongoing source of human genetic diversity. L1/Alu elements are expected to ensure their transmission to subsequent generations by retrotransposing in germ cells or during early embryonic development. Here, we determined that several subfamilies of Alu elements are expressed in undifferentiated human embryonic stem cells (hESCs) and that most expressed Alu elements are active elements. We also exploited expression from the L1 antisense promoter to map expressed elements in hESCs. Remarkably, we found that expressed Alu elements are enriched in the youngest subfamily, Y, and that expressed L1s are mostly located within genes, suggesting an epigenetic control of retrotransposon expression in hESCs. Together, these data suggest that distinct subsets of active L1/Alu elements are expressed in hESCs and that the degree of somatic mosaicism attributable to L1 insertions during early development may be higher than previously anticipated.


Subject(s)
Alu Elements/genetics , Embryonic Stem Cells/physiology , Epigenesis, Genetic , Long Interspersed Nucleotide Elements/genetics , Retroelements/genetics , Animals , Cells, Cultured , Chromosome Mapping , Embryonic Stem Cells/cytology , Female , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Male , Mice , Promoter Regions, Genetic
10.
PLoS One ; 5(11): e15380, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-21085703

ABSTRACT

BACKGROUND: The Saccharomyces cerevisiae RecQ helicase Sgs1 is essential for mitotic and meiotic genome stability. The stage at which Sgs1 acts during meiosis is subject to debate. Cytological experiments showed that a deletion of SGS1 leads to an increase in synapsis initiation complexes and axial associations leading to the proposal that it has an early role in unwinding surplus strand invasion events. Physical studies of recombination intermediates implicate it in the dissolution of double Holliday junctions between sister chromatids. METHODOLOGY/PRINCIPAL FINDINGS: In this work, we observed an increase in meiotic recombination between diverged sequences (homeologous recombination) and an increase in unequal sister chromatid events when SGS1 is deleted. The first of these observations is most consistent with an early role of Sgs1 in unwinding inappropriate strand invasion events while the second is consistent with unwinding or dissolution of recombination intermediates in an Mlh1- and Top3-dependent manner. We also provide data that suggest that Sgs1 is involved in the rejection of 'second strand capture' when sequence divergence is present. Finally, we have identified a novel class of tetrads where non-sister spores (pairs of spores where each contains a centromere marker from a different parent) are inviable. We propose a model for this unusual pattern of viability based on the inability of sgs1 mutants to untangle intertwined chromosomes. Our data suggest that this role of Sgs1 is not dependent on its interaction with Top3. We propose that in the absence of SGS1 chromosomes may sometimes remain entangled at the end of pre-meiotic replication. This, combined with reciprocal crossing over, could lead to physical destruction of the recombined and entangled chromosomes. We hypothesise that Sgs1, acting in concert with the topoisomerase Top2, resolves these structures. CONCLUSIONS: This work provides evidence that Sgs1 interacts with various partner proteins to maintain genome stability throughout meiosis.


Subject(s)
Meiosis/genetics , RecQ Helicases/metabolism , Recombination, Genetic , Saccharomyces cerevisiae Proteins/metabolism , Chromosomes, Fungal/genetics , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Diploidy , Gene Deletion , Genome, Fungal/genetics , Models, Genetic , Protein Binding , RecQ Helicases/genetics , Saccharomyces cerevisiae Proteins/genetics , Sister Chromatid Exchange , Spores, Fungal/genetics
11.
Cell ; 141(7): 1159-70, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20602998

ABSTRACT

Highly active (i.e., "hot") long interspersed element-1 (LINE-1 or L1) sequences comprise the bulk of retrotransposition activity in the human genome; however, the abundance of hot L1s in the human population remains largely unexplored. Here, we used a fosmid-based, paired-end DNA sequencing strategy to identify 68 full-length L1s that are differentially present among individuals but are absent from the human genome reference sequence. The majority of these L1s were highly active in a cultured cell retrotransposition assay. Genotyping 26 elements revealed that two L1s are only found in Africa and that two more are absent from the H952 subset of the Human Genome Diversity Panel. Therefore, these results suggest that hot L1s are more abundant in the human population than previously appreciated, and that ongoing L1 retrotransposition continues to be a major source of interindividual genetic variation.


Subject(s)
Genome, Human , Long Interspersed Nucleotide Elements , Base Sequence , Gene Frequency , Genetics, Population , Humans , Molecular Sequence Data , Phylogeny
12.
Biotechniques ; 46(4): 277-84, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19450234

ABSTRACT

The HeLa cell line is the oldest, most widely distributed, permanent human cell line. As a nearly ubiquitous inhabitant of laboratories using tissue culture techniques, its aggressive growth characteristics make it a problematic contaminant that can overgrow less robust cell lines. Consequently, HeLa contamination is common in both the research laboratory and cell line repository contexts, and its detection is hampered by the lack of a rapid, sensitive and robust assay. Here we report the development of a HeLa-specific DNA diagnostic test: a single duplex detection PCR assay targeting an L1 retrotransposon insertion. All HeLa clones from a geographically diverse panel were positive by this assay, and the particular L1 insertion we identified appears to be unique to the HeLa cell line. The assay can detect very low levels of HeLa contamination (<1%), and can be performed on un-purified cell pellets, allowing rapid routine screening.


Subject(s)
Biomarkers/analysis , Cells, Cultured , HeLa Cells , Long Interspersed Nucleotide Elements/genetics , Equipment Contamination , Gene Frequency , Humans , Polymerase Chain Reaction
13.
Nucleic Acids Res ; 32(13): 3846-55, 2004.
Article in English | MEDLINE | ID: mdl-15272086

ABSTRACT

The initial step in Long Interspersed Element-1 (LINE-1) retrotransposition requires transcription from an internal promoter located within its 5'-untranslated region (5'-UTR). Previous studies have identified a YY1 (Yin Yang 1)-binding site as an important sequence in LINE-1 transcription. Here, we demonstrate that mutations in the YY1-binding site have only minor effects on transcription activation of the full-length 5'-UTR and LINE-1 mobility in a single round cultured cell retrotransposition assay. Instead, these mutations disrupt proper initiation of transcription from the +1 site of the 5'-UTR. Thus, we propose that the YY1-binding site functions as a component of the LINE-1 core promoter to direct accurate transcription initiation. Indeed, this sequence may explain the evolutionary success of LINE-1 by enabling full-length retrotransposed copies to undergo autonomous retrotransposition in subsequent generations.


Subject(s)
Long Interspersed Nucleotide Elements , Transcription Factors/metabolism , Transcription, Genetic , 5' Untranslated Regions , Base Sequence , Binding Sites , Cell Line, Tumor , Erythroid-Specific DNA-Binding Factors , Humans , Molecular Sequence Data , Mutagenesis, Insertional , Mutation , Transcriptional Activation , YY1 Transcription Factor
14.
Proc Natl Acad Sci U S A ; 100(9): 5280-5, 2003 Apr 29.
Article in English | MEDLINE | ID: mdl-12682288

ABSTRACT

Although LINE-1 (long interspersed nucleotide element-1, L1) retrotransposons comprise 17% of the human genome, an exhaustive search of the December 2001 "freeze" of the haploid human genome working draft sequence (95% complete) yielded only 90 L1s with intact ORFs. We demonstrate that 38 of 86 (44%) L1s are polymorphic as to their presence in human populations. We cloned 82 (91%) of the 90 L1s and found that 40 of the 82 (49%) are active in a cultured cell retrotransposition assay. From these data, we predict that there are 80-100 retrotransposition-competent L1s in an average human being. Remarkably, 84% of assayed retrotransposition capability was present in six highly active L1s (hot L1s). By comparison, four of five full-length L1s involved in recent human insertions had retrotransposition activity comparable to the six hot L1s in the human genome working draft sequence. Thus, our data indicate that most L1 retrotransposition in the human population stems from hot L1s, with the remaining elements playing a lesser role in genome plasticity.


Subject(s)
Long Interspersed Nucleotide Elements , Retroelements , Alleles , Gene Frequency , Humans , Open Reading Frames , Phylogeny
15.
Am J Hum Genet ; 72(4): 823-38, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12632328

ABSTRACT

Retrotransposition of L1 LINEs (long interspersed elements) continues to sculpt the human genome. However, because recent insertions are dimorphic, they are not fully represented in sequence databases. Here, we have developed a system, termed "ATLAS" (amplification typing of L1 active subfamilies), that enables the selective amplification and display of DNA fragments containing the termini of human-specific L1s and their respective flanking sequences. We demonstrate that ATLAS is robust and that the resultant display patterns are highly reproducible, segregate in Centre d'Etude du Polymorphisme Humain pedigrees, and provide an individual-specific fingerprint. ATLAS also allows the identification of L1s that are absent from current genome databases, and we show that some of these L1s can retrotranspose at high frequencies in cultured human cells. Finally, we demonstrate that ATLAS also can identify single-nucleotide polymorphisms within a subset of older, primate-specific L1s. Thus, ATLAS provides a simple, high-throughput means to assess genetic variation associated with L1 retrotransposons.


Subject(s)
DNA Transposable Elements/genetics , Long Interspersed Nucleotide Elements/genetics , 5' Untranslated Regions/genetics , DNA/genetics , DNA/isolation & purification , Databases, Factual , Female , Gene Amplification , Humans , Male , Models, Genetic , Molecular Sequence Data , Oligodeoxyribonucleotides , Open Reading Frames , Pedigree , Polymerase Chain Reaction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...