Chromosome-wide identification of novel imprinted genes using microarrays and uniparental disomies (original) (raw)

Journal Article

,

King's College London, School of Medicine at Guy's, King's College and St. Thomas' Hospitals, Department of Medical and Molecular Genetics 8th Floor Guy's Tower, London SE1 9RT, UK

Search for other works by this author on:

,

King's College London, School of Medicine at Guy's, King's College and St. Thomas' Hospitals, Department of Medical and Molecular Genetics 8th Floor Guy's Tower, London SE1 9RT, UK

Search for other works by this author on:

,

King's College London, School of Medicine at Guy's, King's College and St. Thomas' Hospitals, Department of Medical and Molecular Genetics 8th Floor Guy's Tower, London SE1 9RT, UK

Search for other works by this author on:

,

King's College London, School of Medicine at Guy's, King's College and St. Thomas' Hospitals, Department of Medical and Molecular Genetics 8th Floor Guy's Tower, London SE1 9RT, UK

Search for other works by this author on:

,

King's College London, School of Medicine at Guy's, King's College and St. Thomas' Hospitals, Department of Medical and Molecular Genetics 8th Floor Guy's Tower, London SE1 9RT, UK

Search for other works by this author on:

King's College London, School of Medicine at Guy's, King's College and St. Thomas' Hospitals, Department of Medical and Molecular Genetics 8th Floor Guy's Tower, London SE1 9RT, UK

Search for other works by this author on:

Published:

01 January 2006

Revision received:

26 May 2006

Cite

Reiner Schulz, Trevelyan R. Menheniott, Kathryn Woodfine, Andrew J. Wood, Jonathan D. Choi, Rebecca J. Oakey, Chromosome-wide identification of novel imprinted genes using microarrays and uniparental disomies, Nucleic Acids Research, Volume 34, Issue 12, 1 June 2006, Page e88, https://doi.org/10.1093/nar/gkl461
Close

Navbar Search Filter Mobile Enter search term Search

Abstract

Genomic imprinting refers to a specialized form of epigenetic gene regulation whereby the expression of a given allele is dictated by parental origin. Defining the extent and distribution of imprinting across genomes will be crucial for understanding the roles played by imprinting in normal mammalian growth and development. Using mice carrying uniparental disomies or duplications, microarray screening and stringent bioinformatics, we have developed the first large-scale tissue-specific screen for imprinted gene detection. We quantify the stringency of our methodology and relate it to previous non-tissue-specific large-scale studies. We report the identification in mouse of four brain-specific novel paternally expressed transcripts and an additional three genes that show maternal expression in the placenta. The regions of conserved linkage in the human genome are associated with the Prader–Willi Syndrome (PWS) and Beckwith–Wiedemann Syndrome (BWS) where imprinting is known to be a contributing factor. We conclude that large-scale systematic analyses of this genre are necessary for the full impact of genomic imprinting on mammalian gene expression and phenotype to be elucidated.

INTRODUCTION

Genomic imprinting refers to a specialized form of epigenetic gene regulation whereby the expression of a given allele is dictated by its maternal versus paternal origin. Imprinting plays a crucial role in mammalian reproduction and imposes an absolute requirement for maternal and paternal genomes for the generation of viable offspring ( 1 , 2 ). Approximately 80 imprinted genes have been identified in mouse ( http://www.mgu.har.mrc.ac.uk/research/imprinted/ ), and around 40 in human ( 3 ). The extent of imprinting in mouse or human is not fully known, but has been estimated in mouse to range between 100 and 600 genes ( 46 ). Mis-regulation of the imprinted genes has been associated with growth and developmental abnormalities in mice ( 1 , 2 , 4 ), birth defects ( 7 , 8 ) and neoplasias in humans ( 9 ) as well as abnormalities in cloned mammals ( 10 , 11 ).

The rationale for the identification of new imprinted genes is based on further understanding imprinted gene regulation and the genetic components of developmental phenotypes in the mouse and birth defects in humans, such as Prader–Willi Syndrome (PWS), Beckwith–Wiedemann Syndrome (BWS) and cancer. Imprinted genes are frequently associated with asynchronous DNA replication ( 12 ) and epigenetic control mechanisms that can extend over large genomic regions. Studying aspects of epigenetic regulation requires examination of the genomic environment, especially in cases of placenta-specific imprinting where evidence suggests that not only DNA methylation but also histone modification is important for imprinted gene expression ( 13 , 14 ). Thus an inventory of monoallelic expression of genes and transcripts in a given region is advantageous.

Imprinted genes are characteristically differentially expressed in mice with a maternally derived uniparental duplication or disomy (matUpDp/UpD) compared to mice where the same UpDp/UpD is of paternal origin (patUpDp/UpD). Ideally, expression of an imprinted gene transcribed from the maternally inherited allele will increase 2-fold in a matUpDp sample compared to wild-type (wt) and will be undetectable in a patUpDp sample, and vice versa. In contrast, the expression of a non-imprinted gene is not expected to differ. Strains of mice carrying reciprocal and Robertsonian translocation chromosomes have been used to produce progeny where both copies of a particular chromosome or chromosomal region have been inherited from only the mother or the father. Here, the reciprocal translocation mouse strain T65H ( 15 ) was used to generate progeny with UpDps of Chromosomes (Chrs) 7 and 11 either proximal or distal of the translocation breakpoint as described previously ( 16 ). These mice provide the basis for both the computational and molecular studies presented here. They are viable until birth, so that differential gene expression profiles over a range of different tissues were investigated. Robertsonian translocation mouse strains that generated UpDs of Chr 18 ( 17 ) and Chr 12 ( 18 ) were used to gain additional statistical power for the evaluation of this method.

To a limited degree, the effectiveness of this method has already been demonstrated by the identification of a novel brain-specific imprinted gene, Inpp5f_v2 ( 19 ). Here, we report on the results of systematically refining our approach and extending its coverage to the whole of Chr 7 and Chr 11. We describe this methodology, quantitatively evaluate its effectiveness and compare it to previous non-tissue-specific whole genome studies, reviewed in ( 20 ). This study has identified and validated four novel brain-specific paternally expressed transcripts and three placenta-specific maternally expressed genes.

MATERIALS AND METHODS

Tissue sources

Mouse: the tissue sources for the T65H translocation strain are described in ( 16 ). Essentially, newborn mice with maternal and paternal duplications for specific regions of Chrs 7 and 11 were generated using the T65H translocation ( 21 , 22 ). The tissues selected are shown in Table 1 and include 13.5 dpc embryo and placenta, newborn brain, carcass, heart and liver for matUpDp prox 7 versus patUpDp prox 7 (the same samples also generate data for patUpDp prox 11 versus matUpDp prox 11). MatUpDp distal 7 samples were compared to normal sibling samples at 13.5 dpc since the patUpDp distal 7 embryo is not viable at this stage of development. 8.5dpc embryos with maternal versus paternal UpD for Chr 18 were also compared using a Robertsonian translocation Chr Rb(2.8)2Lub(7.18)9Lub or RB92.82Lub (R) and C57BL/6JEi-Rb(7.18)9Lub (B) strains obtained from the Cytogenetic Models Resource at the Jackson Laboratory ( 17 ). Placentae were dissected free from the decidua. However, due to the structure of the placenta with invading maternal vessels, some maternal material is likely to be included in these preparations. 15.5 dpc embryos and placentae from Chr 12 UpD ( 18 ) were obtained in collaboration with Anne Ferguson-Smith. Human: for DHCR7 and AMPD3 , anonymous placenta DNA and RNA samples were collected in collaboration with Dr B.S. Emanuel (CHOP) in accordance with ethical guidelines.

Microarray protocols

Affymetrix Genechips™ U74v2 and 430v2 microarrays were used ( Table 1 ). The U74v2 triple array series represents ∼36 000 genes and ESTs. The U74v2 series represents all sequences (∼6 000) in the Mouse UniGene database (Build 74) that had been functionally characterized at the time plus EST clusters. The 430v2 dual array series probes ∼39 000 genes and ESTs where the 430Av2 array predominantly represents well-characterized genes.

Caesium chloride isolated total RNA from samples in Table 1 were quantified on an Agilent Bioanalyser™ and 5–7 μg was used to prepare biotin-labelled cRNA target essentially as in the Affymetrix™ expression manual ( 23 ). Biotinylated cRNA targets were purified using cRNA Cleanup spin columns (Affymetrix™), fragmented in 5× fragmentation buffer (Affymetrix™), and quantified prior to hybridization on an Agilent Bioanalyser™. A total of 0.15 μg of labelled probe was hybridized per GeneChip expression array followed by washing and staining on the Affymetrix™ fluidics station 450. An Affymetrix Scanner 3000 was used to quantify the signal.

Gene chip operating software (GCOS) data analysis

Data from the scanner were analysed using the Affymetrix™ MASv5 or GCOS software. The software computes the signal for each pair of corresponding PM and MM probes in a probe set as S i = log 2 (PM i ) − log 2 (MM i ) where PM i and MM i are the measured fluorescence levels of the i th probes in the PM and MM sets. In cases where S i would barely exceed the background noise level or be negative, GCOS makes adjustments to ensure a conservative and positive estimate of gene-specific hybridization. GCOS arrives at a single signal value (S) for each probe set by calculating the Tukey-biweight of the S i , a weighted outlier-resistant average. GCOS reports 2 S as the estimate of the absolute expression level of the gene represented by the probe set. GCOS allows a comparative analysis of gene expression in two samples, each used to hybridize a separate microarray. GCOS computes a signal log 2 -ratio (SLR) for each probe set as a measure of the degree of differential expression between the two samples and a P -value as a measure of confidence in any measured difference in expression. In the most common case, the SLR equals the Tukey-biweight over i of log 2 ( A PM i ) − log 2 ( A MM i ) − [log 2 ( B PM i ) − log 2 ( B MM i )]; where A PM i , e.g., is the measured fluorescence of the i th PM probe in a particular probe set on array A (hybridized with sample A). The P -value is based on a Wilcoxon signed rank test applied to the pairs of differences [log 2 ( A PM i ) − log 2 ( A MM i ), log 2 ( B PM i ) − log 2 ( B MM i )] and [log 2 ( A PM i ) − A, log 2 ( B PM i ) − B], where A and B are measures of the background noise of the respective array.

Expression analysis

Two assays were used; the first used cDNA prepared from maternal and paternal UpDp and wt RNA samples in RT–PCR assays. Briefly, 4 μg of total RNA was reverse transcribed using Superscript III (Invitrogen™) reverse transcriptase (manufacturers' instructions) and PCR-amplified using ABgene™ Reddymix and gene-specific primers (Supplementary Table S4). The second method used inter-subspecies hybrid RNA as described in ( 19 ). Essentially, single nucleotide polymorphisms (SNPs) between Mus m. musculus [C57Bl6J (B6)] and Mus m. castaneus (CAST) strains were identified and assayed by DNA sequencing (primers are listed in Supplementary Table S4). RT–PCR followed by sequencing identified the expressing allele(s) for each gene tested (Figures 2 and 3). Sequencing was performed on an ABI 3730 DNA analyser using ABI BigDye™ reagents (manufacturer's protocols). Fetal or parental human DNAs were sequenced for expressed SNPs, which were assayed as described above.

Gene prediction programs

Gene predictions tools used include Ensembl ( 2426 ). SGP ( 27 ), Geneid ( 28 ), Genscan ( 29 ), Superfamily/SCOP ( 30 ), Twinscan ( 31 ) and Augustus ( 32 ).

RESULTS

The ‘one chromosome at a time’ approach excludes most false positives

Each of the microarrays used here measures the expression of thousands of distinct transcripts, all of which can potentially change in expression levels between the respective matUpDp/UpD and patUpDp/UpDs. However, most transcripts can be ruled out based on their location within the genome that places them outside the selected chromosomal duplication or disomy.

Four chromosome anomalies were investigated ( Table 1 ). Filtering by genomic position reduced the number of potential false positives by between 85 and 94% ( Table 2 ). For example, of the 45 037 distinct non-control probe sets, representing over 39 000 transcripts, on the Affymetrix™ 430v2 arrays, only 6595 (14.6%) generate a match to proximal Chrs 7 or 11 when their target sequences are aligned with the genome (March 2005 NCBI build 34) using BLAT ( 33 ). The remaining 38 442 probe sets do not pass the henceforth called ‘map filter’ for this UpDp and thus can be excluded from the search. On average, about a tenth of the transcripts represented on the microarrays mapped to the respective region of UpDp/UpD in our experiments and could legitimately change in expression between the matUpDp/UpD and pat UpDp/UpD samples as a direct result of being imprinted. This reduction of potential false positives by an order of magnitude compared to the analogous whole genome approach (parthenogenote versus androgenote), is compounded by the more limited downstream effects of UpDp/UpDs.

Gains in sensitivity by combining differential expression measures

Arrays hybridized with the matUpDp/UpD sample were compared to arrays hybridized with the patUpDp/UpD sample using the gene operating software (GCOS) that provides two measures of differential expression ( 34 ). The SLR of a probe set captures the fold change in expression of the represented transcript between the two samples, i.e., in simplified terms, SLR = log 2 (expression in matUpDp/UpD / expression in patUpDp/UpD). In addition, GCOS supplies a P -value that expresses the confidence in and direction of any measured expression difference, that is, P = 0.5 if there is no evidence for a change in expression, and P is the closer to 0 or 1 the more statistical evidence there is for an increase or decrease in expression, respectively.

SLR and P -value are closely correlated but non-redundant measures. Frequently, a probe set's absolute SLR (∣SLR∣) is large while 0.5 − ∣P − 0.5∣ ≈ 0.5, meaning that while a large fold change is measured, there is little statistical evidence for differential expression. The converse happens similarly frequently. Which of the two measures more truthfully represents the actual degree of differential expression is not immediately clear. Previously, SLR and P -value have been shown to give qualitatively different results ( 35 ). Here, we systematically evaluated the performance of SLR, P -value and combinations of both, namely, P -value-weighted SLRs (xSLR), in the context of imprinted gene detection. Specifically, xSLR = 0 if 0.5 − ∣P − 0.5∣ ≥ x, and otherwise, xSLR = [1 − 1/x (0.5 − ∣P − 0.5∣)] SLR. Within the interval 0 ≤ 0.5 − ∣P − 0.5∣ ≤ x, xSLR linearly changes from SLR to zero where it remains at all lower levels of statistical confidence. We explored the whole range of possible values for x, i.e. 0 < x ≤ 0.5, and found x = 0.01 (uSLR) to perform best, closely followed by x = 0.05 (vSLR). The simple linear weighing of SLR over the whole range of P -values, (x = 0.5; pSLR) was not nearly as effective.

For each measure, we first estimated from our microarray data, the true positive rate (TPR) and false positive rate (FPR) for a range of differential expression thresholds. The TPR was estimated from a total of 43 known imprinted genes that map to within the investigated UpDp/UpDs and for which there are representative probe sets. For each tissue, we counted (a) the number of genes within the respective UpDp/UpD that the literature has reported to be imprinted in the tissue and for which tissue-matched array data were available and (b) the number of such genes for which in addition, at least one representative probe set detected an increase (for maternally expressed genes) or decrease (for paternally expressed genes) in expression that met or exceeded the differential expression threshold. The TPR was computed as the ratio between the two sums over all tissues of counts (b) and (a). The FPR is the fraction of probe sets mapping to the investigated UpDp/UpDs that do not represent known imprinted genes but nevertheless met or exceeded the differential expression threshold. Plotting the FPR versus the TPR over the range of decision thresholds yields a receiver operating characteristics (ROC) curve shown for SLR, P -value, uSLR, vSLR and pSLR ( Figure 1 ).

A low FPR is paramount to achieve a good cost to discovery ratio in the molecular validation. Even with the map filter, an FPR of 5% still corresponds to, in the proximal Chrs 7 and 11 region, roughly 300 distinct probe sets on the 430 array that will be flagged as representing differentially expressed sequences, but are unlikely to represent imprinted transcripts. We therefore chose 5% as the upper limit on the FPR. Up to this limit, uSLR consistently performed best, i.e. gave the highest TPR compared to the other measures that we considered. Consequently, the ROC curve for uSLR dominates the other curves in Figure 1 for an FPR ≤ 5%. Specifically, the differential expression threshold of ∣uSLR∣ ≥ 0.6 delivered a TPR = 65.8% and an FPR = 4.9% so that henceforth, ‘differentially expressed’ will imply ∣uSLR∣ ≥ 0.6.

The map filter saturates candidates with imprinted genes

To prioritize the molecular validation experiments, we ranked the probe sets passing the map filter by degree of differential expression, i.e. in descending order of ∣uSLR∣. On average, 19% (29%, 38%, 46% and 52%) of all genes known to be imprinted in the respective tissue and mapping to within the respective duplication were ranked among the top 10 (20, 50, 100 and 200). Compared to the analogous ranking of all probe sets, i.e. without the prior application of the map filter, this corresponds to an average increase in the number of known imprinted genes in the top 10 (20, 50, 100 and 200) ranks by 85% (71%, 57%, 61% and 48%). Table 3 provides the absolute values. This illustrates how the map filter increases the saturation of the top ranks with known imprinted genes and, by extrapolation, with likely truely imprinted candidate genes.

Summary statistics of probe sets representing differentially expressed sequences

Table 2 gives a complete account, broken down by UpDp/UpD, tissue and microarray series, of how many probe sets whose target sequences map to within versus to exclusively outside the UpDp/UpD detected differential expression. On average, 4.9% (5.3%) of the probe sets mapping to within (outside) the UpDp/UpD detected differential expression. The differences underlying these averages are not statistically significant ( P = .083; Wilcoxon signed rank test), which suggests that proportionately and in terms of differential expression, a UpDp/UpD affects the region of duplication approximately as much as the rest of the genome.

Differential gene expression profiles across tissues

For the UpDp of proximal Chrs 7 and 11, we conducted separate microarray experiments using placenta, embryo, carcass, brain, liver and heart samples ( Table 1 ). Thus, we created tissue-specific differential expression profiles. Only limited or no tissue-specific data were available for the distal UpDp of Chrs 7 and 11 and the UpDs of Chrs 12 (embryo and placenta) and 18 (embryo).

Profiles of 43 known imprinted genes on Chrs 7, 11, 18 and 12 are compared with the literature-reported imprinting status in Supplementary Table S1. Profiles of the 59 genes on Chrs 7 and 11 that were not known to be imprinted and for which we conducted imprinting validation experiments are listed in Supplementary Table S2, where the results of the validation assays appear side-by-side with the microarray data. The profiles of all transcripts on proximal Chrs 7 and 11 (935 transcripts represented by 1705 probe sets) that were differentially expressed in at least one of the tissues are shown in Supplementary Table S3.

Confirmation of seven differentially expressed candidates

Allele-specific assays using UpDp/UpD RNAs have been used extensively to determine parent of origin specific gene expression ( 36 ), but could be argued not to be independent of the microarray assay since the starting material is the same ( 37 ). Interspecies hybrids have been used to assay for parent of origin specific expression ( 3840 ) but loss of imprinting of some genes has been detected in hybrid crosses ( 41 ). So, where possible we have used more than one method. Moreover, we have tested known imprinted genes and have in every case confirmed imprinting status.

A set of 59 candidate genes with an unknown imprinting status were selected and initially screened in an RT–PCR approach using matUpDp versus patUpDp RNA templates (where available). Candidates showing evidence of parental origin specific expression in this assay on the proximal regions of mouse Chrs 7 were subjected to further validation using RT–PCR combined with a SNP allele-specific assay in mouse inter-sub-specific hybrids.

On proximal Chr 11 in whole embryo or isolated brain, eight candidate imprinted transcripts were tested for monoallelic expression by RT–PCR in mat versus patUpDp material (Supplementary Table S2), along with two control known imprinted genes, U2af1-rs1 and Grb10 . The control genes showed imprinted expression and six out of eight non-control genes were biallelic. Pnpt1 was expressed in paternally duplicated samples only but was not present in wt, Gabra1 showed some maternal bias, but this difference was not considered robust enough to warrant further study. Lsm11 is located within band B1.1 of Chr 11 containing the T65H translocation breakpoint thus may have been duplicated in the distal rather than the proximal experiment. However, the distal microarray data did not suggest differential expression of Lsm11.

On proximal Chr 7, 25 transcripts were tested for monoallelic expression in brain, 17 in whole embryo, 5 in placenta and 2 in heart (Supplementary Table S2). By UpDp assay, 10 transcripts showed allele bias in brain and were tested further by SNP analysis, which confirmed that four of these (BB077283, BM117114, AK080843 and AV328498) were paternally expressed ( Figure 2 ). BB182944 and BB312372 were also validated as paternally expressed but were subsequently found to correspond to the imprinted Pec2 and Pec3 transcripts ( 42 ) and were excluded. Of the remainder, Ampd3 was maternally expressed in placenta. The placenta UpDp assay showed a maternal bias with some paternal expression (data not shown) rather than the exclusive maternal expression seen with the interspecies SNP assay ( Figure 3 ). BB264453 was robustly differentially expressed by RT–PCR in mat versus pat UpDp brain, but did not contain a SNP between B6 and cast and so was not considered further. The location of AI114950 within band F4 of Chr 7 makes it analogous to Lsm11 and the distal microarray data did not suggest differential expression of AI114950. The remaining tested transcripts were biallelic by SNP assay. AK080843 and AV328498 are transcribed in the antisense orientation relative to Ube3A and could be part of the large imprinted antisense transcript LNCAT, ( 43 , 44 ) but because the LNCAT cDNA is not clearly defined or publicly available, this is uncertain.

Ten transcripts were selected from distal mouse Chr 7 (Supplementary Table S2). Of these, Th and Dhcr7 were maternally expressed in placenta ( Figure 3 ) but were biallelic in embryo. These transcripts were not as robustly monoallelic as the novel brain transcripts in the proximal region, although the allele preferences exchanged with reversed parental transmission ( Figure 3 ).

Verified imprinted ESTs: gene predictions and genomic context

Gene predictors were applied to the genomic regions containing the ESTs for which validation by RT–PCR and SNP analysis had confirmed imprinting ( Figure 2 ). The genomic position of each EST was uniquely identifiable. In brain and embryo, the paternally expressed AK080843 and AV328498 map to a region between 6.7 and 13 kb centromeric of Snrpn ( Figure 2A ). Based on the genomic sequence of this region, no prediction program provided evidence for a separate gene or an extended transcript of Snrpn whose coding region aligns with the ESTs. AV328498 is the 3′-read of the full insert sequence AK078094 that partially overlaps AK080843. Both AK078094 and AK080843 partially overlap BC070450, a transcript extending much further 3′. The distinct splicing patterns of these transcripts suggest that they might be alternative transcripts of the same transcriptional unit. Whether these transcripts are extensions of Snrpn is not completely clear but based on the ESTs in the region (including AK080843 and AV328498, AK078094), Ensembl predicts a gene (ENSMUSG00000016158) that is distinct from Snrpn at this location. To further ascertain whether transcripts AV328498 and AK080843 could splice onto Snrpn , a Snrpn forward primer was combined with the AV328498 and AK080843 reverse primers (Supplementary Table S4, SNP assay primers) to amplify brain cDNA by RT–PCR. ABGene thermoprime plus Taq polymerase was used to amplify large products of up to 12 kb. No products were seen from any primer combination (data not shown). This provides empirical data to support these transcripts being independent of Snrpn , but they are limited in assaying the absence of a product. Similarly, no genes were predicted that coincide with BB077283 (corresponding 5′-read: BB625859) or BM117114 (no 5′-read available, but identical to BQ555876 with corresponding 5′-read BQ555877). These ESTs map telomeric of Snrpn and centromeric of Ndn ( Figure 2A ) and are located distant (1.8 Mb; BM117114 and 1.3 Mb; BB077283) from Snrpn and hence are not likely to be part of the Snrpn transcript.

Human orthologues of mouse distal Chromosome 7 genes are not maternally expressed in placenta

We examined the expression status of the human orthologues DHCR7 and AMPD3 in placenta using a combined RT–PCR and SNP assay essentially as performed in the mouse tissues. Both genes were biallelically expressed in human term placentae ( Figure 4 ). The imprinting status of TH was not addressed, since informative polymorphisms could not be found in our sample sets. These genes may however be imprinted in other non-tested human tissues since tissue-specific imprinting need not be the same between species ( 45 ).

Limited overlap with previous non-tissue-specific whole genome imprinting studies

A 2-fold change in expression was used as the cut-off for differential expression in RIKEN's FANTOM2 imprinting screen ( 46 ), which identified 2110 imprinting candidate transcripts. A large fraction of these transcripts were represented by probe sets on the Affymetrix™ arrays (430v2: 79%, U74v2: 65%), and between 17 (350) and 41% (870) of them were represented by probe sets mapping to one of the investigated UpDp/UpDs. Given that the above transcripts constitute imprinting candidates, we expected a large fraction of them to be represented by one or more differentially expressed probe sets in our experiments. However, at the most, this fraction was 45% (UpD of Chr 18), and across all UpDp/UpDs, tissues and microarray series', the average was 20.4%. The overlap between the RIKEN imprinting candidates and differentially expressed probe sets in our experiments increased when a less stringent differential expression threshold was used, permitting a higher FPR. For example, using ∣pSLR∣ ≥ 0.5 with a TPR of 74% and an FPR of 21.1% ( Figure 1 ), the overlap increases to 66.6% in the best case (UpD of Chr 18), and to 50.3% on average. This suggests that the ∣uSLR∣ ≥ 0.6 is more stringent than the 2-fold change in expression threshold used in the RIKEN study.

Recent work that used bioinformatic methods predicted 600 out of 23 788 annotated (Ensembl) autosomal mouse genes to be imprinted based on their similarity to sequence features surrounding 44 known imprinted genes, and their dissimilarity to ∼500 assumed non-imprinted genes ( 6 ). The TPR and FPR of the analysis, determined by cross-validation, was 100 and 7%, respectively. Using the same method, the authors also predicted allele preference, that cross-validation showed to be 97.7% accurate. We observed the largest overlap with our study on proximal Chrs 7 and 11 to which 27 of the 600 predicted imprinted genes mapped and were represented by probe sets on the Affymetrix arrays. For 10 of these genes there was at least one probe set that detected differential expression in at least one of the investigated tissues, and in three cases, the predicted allele preference consistently agreed with the direction of change detected by the probe sets (Supplementary Table S5).

DISCUSSION

Microarray measurements versus allele-specific assays

In principle, microarrays are an ideal tool for the large-scale detection of imprinted genes in UpDp/UpD material because imprinted genes are expected to exhibit an extreme expression differential between the matUpDp/UpD and patUpDp/UpD samples. Empirically however, the array measurements will lead to both false negatives and false positives due to, among other factors, the sharing of probe sets between multiple alternative transcripts, cross-hybridization and downstream effects of the chromosome anomaly. Most downstream targets can be excluded by map position. However some false positives are retained, and unless a gene has been verified by allele-specific assay, its imprinting status remains uncertain. Thus, the fine-tuning of the true and especially the FPR in microarray data analysis is of paramount importance in a screen, such as this and has been a focus of this study.

Probe sets representing differentially expressed sequences (∣uSLR∣ ≥ .6) were present for 40 of the 59 genes that were tested for imprinting using the UpDp and/or the SNP assay. Nineteen transcripts were assayed based on past analyses using outdated Affymetrix™ software and annotation (MAS4). For individual tissues, Table 4 shows the total number of transcripts for individual tissues that were tested using an allele-specific assay and compares the results with the microarray measurements. The control gene, H19 , showed robust maternal expression in the SNP assay ( Figure 3 ). However, contrary to expectation, on examining the differential expression on the embryo and placenta distal matUpDp versus wt microarrays, H19 had similar expression levels in both samples (Supplementary Table S1). In contrast, Igf2 , was expressed in wt and vastly reduced in matUpDp RNA as would be predicted (Supplementary Table S1). The probe sequences for H19 were examined but there was no evidence for cross-hybridization in the Affymetrix™ annotation. Since H19 is a very highly expressed transcript in the cell, the fluorescent signal may have saturated, but this was not the case because GCOS excludes saturated readings from the comparative analysis, and for H19 , all probes were included. Hence this false negative result may be an artifactual problem, but could also be due to a biological modulation of H19 RNA levels in the matUpDp samples by an unknown mechanism.

More than 66% of the 40 genes with microarray evidence for differential expression were found to be biallelic in the UpDp and/or SNP assays ( Table 4 ). This is in stark contrast to the FPR of 5% that we estimated for our threshold for differential expression (∣uSLR∣ ≥ .6; Figure 1 ). However, this estimate is with respect to differential expression detected by a single probe set. Each of the tested 59 genes is on average represented by four probe sets that were each used for measurements in at least two different tissues. Assuming independence, the probability of one of these probe sets detecting differential expression in one of the tissues by chance is roughly one-third. However, only 9 of the 31 genes detected as differentially expressed by their probe sets subsequently exhibited an allele preference in the UpDp assay. For a per-gene and cross-tissue FPR of 1/3 the expected number is 20. The UpDp assay is not strand-specific, so an antisense transcript could mask an allele bias in the UpDp assay, but this is unlikely to explain the ∼50% shortfall in genes with an allele bias. The probe sets that apparently mistakenly indicate differential expression do not obviously share any characteristics that distinguish them from ‘correct’ probe sets. So, a large fraction of the inconsistencies between the microarray and the UpDp assay results remain unexplained.

Limited overlap with previous whole genome imprinting studies

Previous studies report that the genome-wide identification of imprinted genes via differential expression between parthenogenotes and androgenotes likely suffers from a high FPR due to the extreme and different downstream effects of partheno- and androgenesis ( 39 , 47 ). This is likely to explain our limited overlap with the results of the RIKEN FANTOM2 imprinting study ( 46 ), especially since relaxing our threshold for differential expression increased the overlap significantly.

The small overlap between our results and the genes predicted to be imprinted in ( 6 ) has several possible explanations. The predictor may have misclassified imprinted genes for which the training set was not representative, e.g. imprinted genes with a distinct regulatory mechanism that would have made the sequence features appear atypical for an imprinted gene. For a given gene prediction, it is still unknown in which tissue(s) and developmental stages the imprinting occurs. Our microarray measurements may not have covered the relevant tissues or developmental stages. A general difficulty in establishing a closer correspondence between the classifier and our microarray-based approach is the classifiers' limited applicability to characterized genes, while the microarrays contain a large number of EST-complementary probe sets.

Maternally expressed genes in placenta

Two of the three genes found to be maternally expressed in the placenta, Dhcr7 and Th are located within or in close association with the BWS orthologous region on mouse distal Chr 7. The third, Ampd3 , is not associated with the cluster and maps ∼33 Mb centromeric of H19 in apparent isolation from any other imprinted gene. The majority of genes contained within the respective human and mouse BWS regions are highly conserved, both in structural organization and, with few exceptions, in imprinting status ( 48 , 49 ). We could not address the imprinting status of TH in humans due to the absence of informative polymorphisms in the available tissues. DHCR7 and AMPD3 were biallelic in placenta, a finding consistent with their respective locations on 11q13 and 11p15.4, neither region being associated with imprinting ( Figure 4 ).

The boundaries of the cluster have been defined by the maternally expressed H19 and Osbpl5 genes ( 4850 ). The finding of preferential maternal expression at Dhcr7 , which is located outside these arbitrary boundaries could extend this region of imprinting. Current evidence supports regional imprinting control in the BWS cluster by two imprinting centres (IC's), the H19 differentially methylated domain (DMD) ( 51 , 52 ), and the Kcnq1 (Kv) DMR1 ( 40 , 53 ). Paternal inheritance of a KvDMR1 deletion leads to loss of imprinting of several maternally expressed imprinted genes with disruption of the repressive paternally expressed Kcnq1ot1 antisense transcript thought to be responsible for this effect. Though not formally addressed here, it is conceivable that the maternal expression of Th and Dhcr7 , both of which are located adjacent to KvDMR1 could be regulated by this region, or by the Kcnq1ot1 transcript in cis . Analysis of both genes within the context of a KvDMR1 deletion allele could address this expectation.

One of the limitations of examining monoallelic expression in placenta compared to other organs is the presence of invading maternal blood vessels. The allele-specific assay in interspecies hybrids were performed on placenta with the deciduum removed but the precise fetal:maternal contribution is not known. Some maternal expression could be derived from maternal contamination. The maternal and paternal UPD placentae would not be susceptible to this issue since it applies equally to these tissues harvested for the arrays. Thus the arrays provide independent evidence for differential expression. Maternal allele preference observed in these genes is largely consistent with the directionality of imprinting of other known placental imprinted genes, an exception provided by the Igf2 P0 transcript, expressed exclusively from the paternal allele in labyrinthine trophoblast ( 54 , 55 ).

While Ampd3 shows almost exclusive expression from the maternal allele, for Dhcr7 and Th , transcription was also evident (albeit at a much lower level) from the paternal allele, which indicates that maternal expression of these genes may not be absolute. On the other hand we note the caveat of alternative (non-imprinted) transcripts or imprinting in a cell lineage-dependent manner that potentially complicates this interpretation. However it has not escaped our notice that similarly ‘incomplete’ imprinting has been observed for other placenta-specific genes, Nap1l4, Phlda2 and Osbpl5 , located immediately centromeric to Dhcr7 ( 48 , 49 ). Incomplete silencing of the paternal allele in these examples, it has been argued, reflects their relatively distant separation from the KvDMR1 compared with other genes, Kcnq1, Kcnq1ot1 and Cdkn1c , that are more closely associated with this element and robustly imprinted ( 49 ), providing a possible mechanistic explanation for these observations.

Imprinted EST transcripts identified in the PWS/Angelman Syndrome region

PWS is thought to arise as a consequence of the loss in expression of several paternally expressed imprinted genes on human 15q11–13. Studies of transgenic models have not revealed an obvious candidate, though the Ndn gene, when disrupted in mice, causes failure to thrive, a frequent observation in PWS ( 56 ). The characterization of additional imprinted transcripts within this region could therefore contribute to the further genetic dissection of PWS. Four novel imprinted transcripts AK080843, BM117114, BB077283 and AV328498 were identified in the PWS/AS orthologous region on proximal Chr 7. Bioinformatic analysis did not reveal evidence that these transcripts were contained within larger transcription units or possessed a capacity for protein coding. Significantly, human orthologues could not be found for two transcripts ( BM117114 and BB077283 ), suggesting that they do not have a role in PWS. For AK080843 , a 100 bp sequence shares 96% identity in the human genome although extensive RT–PCR analysis failed to detect transcripts from this region (T. R. Menheniott, unpublished data).

Unifying hypotheses to explain imprinting disorders will require a comprehensive mapping of genes in the pertinent critical regions. Methods permitting the global detection of imprinted genes across multiple developmental lineages are likely to shed light upon the role of imprinting processes in such disorders. Indeed a prominent conclusion of this study is that the total number of imprinted genes is likely to exceed the number of currently known imprinted genes and the incidence of tissue-specific imprinting will be significant.

Table 1

Overview of the microarray data used in this study

UpDp/UpD Microarray Tissue
Series Model Pl Em Cc Br Lr Ht
Prox Chrs 7 and 11 Affymetrix™ A x x
430v2 B x x
Affymetrix™ A x x x x
U74v2 B x x x x
C x x x
Incyte™ GEM 1 x x x
Dist Chrs 7 and 11 Affymetrix™ A x x
U74v2 B x x
C x x
Chr 12 Affymetrix™ A x x
U74v2 B x x
C x x
Chr 18 Affymetrix™ A x
430v2 B x
UpDp/UpD Microarray Tissue
Series Model Pl Em Cc Br Lr Ht
Prox Chrs 7 and 11 Affymetrix™ A x x
430v2 B x x
Affymetrix™ A x x x x
U74v2 B x x x x
C x x x
Incyte™ GEM 1 x x x
Dist Chrs 7 and 11 Affymetrix™ A x x
U74v2 B x x
C x x
Chr 12 Affymetrix™ A x x
U74v2 B x x
C x x
Chr 18 Affymetrix™ A x
430v2 B x

An ‘x’ indicates the availability of data for a specific combination of UpDp/UpD, microarray and tissue (Pl = placenta, Em = embryo, Cc = carcass, Br = brain, Lv = liver and Ht = heart). Note that for the experiment using a prox Chrs 7 and 11 sample of heart, there was only enough material to hybridize the U74v2 A and B arrays.

Table 1

Overview of the microarray data used in this study

UpDp/UpD Microarray Tissue
Series Model Pl Em Cc Br Lr Ht
Prox Chrs 7 and 11 Affymetrix™ A x x
430v2 B x x
Affymetrix™ A x x x x
U74v2 B x x x x
C x x x
Incyte™ GEM 1 x x x
Dist Chrs 7 and 11 Affymetrix™ A x x
U74v2 B x x
C x x
Chr 12 Affymetrix™ A x x
U74v2 B x x
C x x
Chr 18 Affymetrix™ A x
430v2 B x
UpDp/UpD Microarray Tissue
Series Model Pl Em Cc Br Lr Ht
Prox Chrs 7 and 11 Affymetrix™ A x x
430v2 B x x
Affymetrix™ A x x x x
U74v2 B x x x x
C x x x
Incyte™ GEM 1 x x x
Dist Chrs 7 and 11 Affymetrix™ A x x
U74v2 B x x
C x x
Chr 12 Affymetrix™ A x x
U74v2 B x x
C x x
Chr 18 Affymetrix™ A x
430v2 B x

An ‘x’ indicates the availability of data for a specific combination of UpDp/UpD, microarray and tissue (Pl = placenta, Em = embryo, Cc = carcass, Br = brain, Lv = liver and Ht = heart). Note that for the experiment using a prox Chrs 7 and 11 sample of heart, there was only enough material to hybridize the U74v2 A and B arrays.

Table 2

For each combination of microarray series, UpDp/UpD and tissue (Pl = placenta, Em = embryo, Cc = carcass, Br = brain, Lv = liver and Ht = heart) used in this study, the table shows the number of distinct non-control probe sets detecting differential expression that map to within (normal script) versus to only outside ( italic script) the UpDp/UpD

UpDp/UpD: Prox Chrs 7 and 11 Dist Chrs 7 and 11 Chr 12 Chr 18
Array series Number of distinct non-control probe sets
Mapping to within (normal) verus to only outside (italic) the UpDp/UpD
Total Total luSLR| ≥ 6 Total luSLR| ≥ 6 Total luSLR| ≥ 6 Total luSLR| ≥ 6
Pl Em Cc Br Lv Ht Pl Em Pl Em Em
430 45037 6595 222 212 3852 245
38442 1321 1084 41185 2246
U74 36701 3679 161 305 325 125 3157 107 101 2240 58 267
33022 1227 2451 2650 854 33544 1128 1321 34461 603 4369
UpDp/UpD: Prox Chrs 7 and 11 Dist Chrs 7 and 11 Chr 12 Chr 18
Array series Number of distinct non-control probe sets
Mapping to within (normal) verus to only outside (italic) the UpDp/UpD
Total Total luSLR| ≥ 6 Total luSLR| ≥ 6 Total luSLR| ≥ 6 Total luSLR| ≥ 6
Pl Em Cc Br Lv Ht Pl Em Pl Em Em
430 45037 6595 222 212 3852 245
38442 1321 1084 41185 2246
U74 36701 3679 161 305 325 125 3157 107 101 2240 58 267
33022 1227 2451 2650 854 33544 1128 1321 34461 603 4369

The table also provides the total number of non-control probe sets per microarray series and, for each UpDp/UpD in turn, the total number of probe sets that map to within versus to only outside the UpDp/UpD.

Table 2

For each combination of microarray series, UpDp/UpD and tissue (Pl = placenta, Em = embryo, Cc = carcass, Br = brain, Lv = liver and Ht = heart) used in this study, the table shows the number of distinct non-control probe sets detecting differential expression that map to within (normal script) versus to only outside ( italic script) the UpDp/UpD

UpDp/UpD: Prox Chrs 7 and 11 Dist Chrs 7 and 11 Chr 12 Chr 18
Array series Number of distinct non-control probe sets
Mapping to within (normal) verus to only outside (italic) the UpDp/UpD
Total Total luSLR| ≥ 6 Total luSLR| ≥ 6 Total luSLR| ≥ 6 Total luSLR| ≥ 6
Pl Em Cc Br Lv Ht Pl Em Pl Em Em
430 45037 6595 222 212 3852 245
38442 1321 1084 41185 2246
U74 36701 3679 161 305 325 125 3157 107 101 2240 58 267
33022 1227 2451 2650 854 33544 1128 1321 34461 603 4369
UpDp/UpD: Prox Chrs 7 and 11 Dist Chrs 7 and 11 Chr 12 Chr 18
Array series Number of distinct non-control probe sets
Mapping to within (normal) verus to only outside (italic) the UpDp/UpD
Total Total luSLR| ≥ 6 Total luSLR| ≥ 6 Total luSLR| ≥ 6 Total luSLR| ≥ 6
Pl Em Cc Br Lv Ht Pl Em Pl Em Em
430 45037 6595 222 212 3852 245
38442 1321 1084 41185 2246
U74 36701 3679 161 305 325 125 3157 107 101 2240 58 267
33022 1227 2451 2650 854 33544 1128 1321 34461 603 4369

The table also provides the total number of non-control probe sets per microarray series and, for each UpDp/UpD in turn, the total number of probe sets that map to within versus to only outside the UpDp/UpD.

 ROC curves for the SLR (solid), P -value (dashed), pSLR (finely dashed), uSLR (dotted) and vSLR (dash-dotted) differential expression measures. Each curve shows the TPR y -axis in relation to the FPR x -axis, estimated at various different decision thresholds for the respective measure and linearly interpolated in between the estimates. Labelled arrows point out thresholds of particular importance, specifically, the thresholds at which the estimated FPR was roughly 5% for all five measures, and/or, for uSLR and vSLR, thresholds that marked the end of steep increases in the TPR and therefore constituted particularly good trade-offs between TPR and FPR. The legend in the figure states for each measure the condition that a probe set needed to satisfy in order to be considered differentially expressed where r, s, t, u and v are the decision thresholds, and P and SLR are the P -value and SLR as computed by GCOS. d denotes Kronecker-δ, that equals 1 if the boolean expression in parenthesis (in our case unequality) is true, and that equals 0 otherwise. The uSLR consistently achieved the highest TPR for up to an FPR of 5%, closely followed by vSLR. Most significantly, P -value and SLR in isolation performed much worse as measures of differential expression than when combined in measures like uSLR.

Figure 1

ROC curves for the SLR (solid), P -value (dashed), pSLR (finely dashed), uSLR (dotted) and vSLR (dash-dotted) differential expression measures. Each curve shows the TPR y -axis in relation to the FPR x -axis, estimated at various different decision thresholds for the respective measure and linearly interpolated in between the estimates. Labelled arrows point out thresholds of particular importance, specifically, the thresholds at which the estimated FPR was roughly 5% for all five measures, and/or, for uSLR and vSLR, thresholds that marked the end of steep increases in the TPR and therefore constituted particularly good trade-offs between TPR and FPR. The legend in the figure states for each measure the condition that a probe set needed to satisfy in order to be considered differentially expressed where r, s, t, u and v are the decision thresholds, and P and SLR are the P -value and SLR as computed by GCOS. d denotes Kronecker-δ, that equals 1 if the boolean expression in parenthesis (in our case unequality) is true, and that equals 0 otherwise. The uSLR consistently achieved the highest TPR for up to an FPR of 5%, closely followed by vSLR. Most significantly, P -value and SLR in isolation performed much worse as measures of differential expression than when combined in measures like uSLR.

Table 3

For each combination of tissue (Pl = placenta, Em = embryo, Cc = carcass, Br = brain, Lv = liver and Ht = heart) and microarray series, the table first shows the union of the respectively studied UpDp/UpDs, then the total number of known imprinted genes located within the corresponding genomic regions, and finally, the number of known imprinted genes for which probe sets ranked in the top 10, 20, 50, 100 and 200, respectively, when the probe sets on all arrays of the series were arranged in descending order of |uSLR|, with (normal script) and without ( italic script) prior application of the map filter.

Tissue Array series Union of UpDp/UpDs Number of known imprinted genes
Total Ranked in top …
10 20 50 100 200
Br 430 Prox Chrs 7 and 11 20 4 6 10 11 13
3 6 7 9 11
Cc U74 Prox Chrs 7 and 11 12 4 5 6 6 7
2 3 4 5 6
Em 430 Prox Chrs 7 and 11, Chr 18 21 3 7 8 12 12
3 6 7 7 9
U74 Dist Chrs 7 and 11, Chr 12 19 3 4 4 5 6
1 1 2 3 3
Ht U74 Prox Chrs 7 and 11 10 4 5 5 6 6
2 3 4 5 5
Lv U74 Prox Chrs 7 and 11 12 2 2 3 5 5
1 1 2 2 3
Pl U74 Chrs 7 and 11, Chr 12 31 4 7 11 13 16
1 1 4 5 7
Tissue Array series Union of UpDp/UpDs Number of known imprinted genes
Total Ranked in top …
10 20 50 100 200
Br 430 Prox Chrs 7 and 11 20 4 6 10 11 13
3 6 7 9 11
Cc U74 Prox Chrs 7 and 11 12 4 5 6 6 7
2 3 4 5 6
Em 430 Prox Chrs 7 and 11, Chr 18 21 3 7 8 12 12
3 6 7 7 9
U74 Dist Chrs 7 and 11, Chr 12 19 3 4 4 5 6
1 1 2 3 3
Ht U74 Prox Chrs 7 and 11 10 4 5 5 6 6
2 3 4 5 5
Lv U74 Prox Chrs 7 and 11 12 2 2 3 5 5
1 1 2 2 3
Pl U74 Chrs 7 and 11, Chr 12 31 4 7 11 13 16
1 1 4 5 7

Table 3

For each combination of tissue (Pl = placenta, Em = embryo, Cc = carcass, Br = brain, Lv = liver and Ht = heart) and microarray series, the table first shows the union of the respectively studied UpDp/UpDs, then the total number of known imprinted genes located within the corresponding genomic regions, and finally, the number of known imprinted genes for which probe sets ranked in the top 10, 20, 50, 100 and 200, respectively, when the probe sets on all arrays of the series were arranged in descending order of |uSLR|, with (normal script) and without ( italic script) prior application of the map filter.

Tissue Array series Union of UpDp/UpDs Number of known imprinted genes
Total Ranked in top …
10 20 50 100 200
Br 430 Prox Chrs 7 and 11 20 4 6 10 11 13
3 6 7 9 11
Cc U74 Prox Chrs 7 and 11 12 4 5 6 6 7
2 3 4 5 6
Em 430 Prox Chrs 7 and 11, Chr 18 21 3 7 8 12 12
3 6 7 7 9
U74 Dist Chrs 7 and 11, Chr 12 19 3 4 4 5 6
1 1 2 3 3
Ht U74 Prox Chrs 7 and 11 10 4 5 5 6 6
2 3 4 5 5
Lv U74 Prox Chrs 7 and 11 12 2 2 3 5 5
1 1 2 2 3
Pl U74 Chrs 7 and 11, Chr 12 31 4 7 11 13 16
1 1 4 5 7
Tissue Array series Union of UpDp/UpDs Number of known imprinted genes
Total Ranked in top …
10 20 50 100 200
Br 430 Prox Chrs 7 and 11 20 4 6 10 11 13
3 6 7 9 11
Cc U74 Prox Chrs 7 and 11 12 4 5 6 6 7
2 3 4 5 6
Em 430 Prox Chrs 7 and 11, Chr 18 21 3 7 8 12 12
3 6 7 7 9
U74 Dist Chrs 7 and 11, Chr 12 19 3 4 4 5 6
1 1 2 3 3
Ht U74 Prox Chrs 7 and 11 10 4 5 5 6 6
2 3 4 5 5
Lv U74 Prox Chrs 7 and 11 12 2 2 3 5 5
1 1 2 2 3
Pl U74 Chrs 7 and 11, Chr 12 31 4 7 11 13 16
1 1 4 5 7

 Identification of novel paternally expressed transcripts on mouse proximal Chr 7. ( A ) The 2 Mb PWS/AS orthologous region with positions of the novel paternally expressed transcripts indicated * . Genes are shown as open boxes with the relative transcriptional orientations defined by arrows. Blue, red or no colouring define paternal, maternal or biallelic expression respectively. ( B ) RT–PCR analysis of candidates identified on the proximal Chr 7 brain array in cDNA derived from patDp prox 7, matDp prox 7 and wild-type brain tissue. Controls for paternal ( Snrpn ), maternal ( Grb10 ) and biallelic ( Igf1r ) expression are shown. The molecular weight marker is a 100 bp DNA ladder where the bright band corresponds to 500 bp. Samples treated with and without reverse transcriptase are indicated as + or −RT. ESTs AK080843, BB077283, BM117114 and AV328498 were paternally expressed. ( C ) Allele-specific RT–PCR analysis of proximal Chr 7 candidates. Newborn brain tissues with expressed SNPs were obtained from reciprocal crosses between M.m.musculus (B6) and M.m.castaneus (CAST) animals. cDNA fragments containing the SNPs were recovered by RT–PCR and direct sequencing to determine allele-specific expression.

Figure 2

Identification of novel paternally expressed transcripts on mouse proximal Chr 7. ( A ) The 2 Mb PWS/AS orthologous region with positions of the novel paternally expressed transcripts indicated * . Genes are shown as open boxes with the relative transcriptional orientations defined by arrows. Blue, red or no colouring define paternal, maternal or biallelic expression respectively. ( B ) RT–PCR analysis of candidates identified on the proximal Chr 7 brain array in cDNA derived from patDp prox 7, matDp prox 7 and wild-type brain tissue. Controls for paternal ( Snrpn ), maternal ( Grb10 ) and biallelic ( Igf1r ) expression are shown. The molecular weight marker is a 100 bp DNA ladder where the bright band corresponds to 500 bp. Samples treated with and without reverse transcriptase are indicated as + or −RT. ESTs AK080843, BB077283, BM117114 and AV328498 were paternally expressed. ( C ) Allele-specific RT–PCR analysis of proximal Chr 7 candidates. Newborn brain tissues with expressed SNPs were obtained from reciprocal crosses between M.m.musculus (B6) and M.m.castaneus (CAST) animals. cDNA fragments containing the SNPs were recovered by RT–PCR and direct sequencing to determine allele-specific expression.

 Identification of novel maternally expressed transcripts on mouse distal Chr 7. ( A ) The ∼1 Mb BWS orthologous region shown with positions of the novel maternally expressed genes indicated * . Regions of conserved linkage on human chromosomes are indicated by horizontal bars. Genes are shown as open boxes with the relative transcriptional orientations defined by arrows. Blue, red or no colouring define paternal, maternal or biallelic expression respectively. ( B ) Allele-specific RT–PCR analysis of distal Chr 7 candidates. Embryo and placenta (E13.5) tissues with expressed SNPs were obtained by performing reciprocal crosses between M.m.musculus (B6) and M.m.castaneus (CAST) animals. cDNA fragments containing the SNPs were recovered by RT–PCR then direct sequenced to determine allele-specific expression. Distal Chr 7 candidates; Dhcr7, Th and Ampd3 were maternally expressed in E13.5 placenta (lower panels) but biallelic in E13.5 embryo (middle panels). Ubiquitously imprinted ( H19 ) and biallelic ( Tnnt3 ) control genes are shown.

Figure 3

Identification of novel maternally expressed transcripts on mouse distal Chr 7. ( A ) The ∼1 Mb BWS orthologous region shown with positions of the novel maternally expressed genes indicated * . Regions of conserved linkage on human chromosomes are indicated by horizontal bars. Genes are shown as open boxes with the relative transcriptional orientations defined by arrows. Blue, red or no colouring define paternal, maternal or biallelic expression respectively. ( B ) Allele-specific RT–PCR analysis of distal Chr 7 candidates. Embryo and placenta (E13.5) tissues with expressed SNPs were obtained by performing reciprocal crosses between M.m.musculus (B6) and M.m.castaneus (CAST) animals. cDNA fragments containing the SNPs were recovered by RT–PCR then direct sequenced to determine allele-specific expression. Distal Chr 7 candidates; Dhcr7, Th and Ampd3 were maternally expressed in E13.5 placenta (lower panels) but biallelic in E13.5 embryo (middle panels). Ubiquitously imprinted ( H19 ) and biallelic ( Tnnt3 ) control genes are shown.

 Imprinting analysis in human placenta. For DHCR7 two distinct polymorphisms, a G/A SNP at nucleotide 364 and a known T/C SNP at nucleotide 382 (rs1790334) in the DHCR7 cDNA were identified in two individuals by sequencing fetal DNA samples. For the AMPD3 analysis a T/A SNP at nucleotide 3160 in the AMPD3 cDNA was identified in one individual by sequencing fetal DNA samples. Allele-specific expression analysis in matched placenta cDNA showed the genes to be biallelic.

Figure 4

Imprinting analysis in human placenta. For DHCR7 two distinct polymorphisms, a G/A SNP at nucleotide 364 and a known T/C SNP at nucleotide 382 (rs1790334) in the DHCR7 cDNA were identified in two individuals by sequencing fetal DNA samples. For the AMPD3 analysis a T/A SNP at nucleotide 3160 in the AMPD3 cDNA was identified in one individual by sequencing fetal DNA samples. Allele-specific expression analysis in matched placenta cDNA showed the genes to be biallelic.

Table 4

Contingency table summarizing Supplementary Table S2 by contrasting the microarray-derived mode of expression (either M = maternal: uSLR ≥ .6, P = paternal: uSLR ≤ −.6 or B = biallelic) with the mode determined by the UpDp/UpD PCR and/or SNP sequencing assays

Mode of expression based on microarray measurement
Array-based Maternal Paternal Biallelic
expression mode: l M P B M P B M P B
Tissue Total
Pl 13 2 2 9
Ampd3 Dhcr7
Tspan4 Th
Em 28 9 2 4 13
A330103N21Rik AK080843 Ebf3
Ccndl BB264453 Gabra5
Ceacam11-14 Hbxap
Cebpa Rtn4
Pank3
Teadl
Br 30 1 5 8 11 5
Gabral* Rkhd3 Inpp5f_v2 Ebf2
Rgs10 AK080843 Txnl2
MGI:2446326 BM117114 Mlstd2
Arrdc4 BB312372* Kcnq5
0710005119Rik BB264453* Dlgh2
BB182944 A1114950
BB077283 AA123443
AV328498 4933439C20Rik
4632427E13Rik
4632419K20Rik
2310047115Rik
Ht 2 1 1
Sh3gl3
Mode of expression based on microarray measurement
Array-based Maternal Paternal Biallelic
expression mode: l M P B M P B M P B
Tissue Total
Pl 13 2 2 9
Ampd3 Dhcr7
Tspan4 Th
Em 28 9 2 4 13
A330103N21Rik AK080843 Ebf3
Ccndl BB264453 Gabra5
Ceacam11-14 Hbxap
Cebpa Rtn4
Pank3
Teadl
Br 30 1 5 8 11 5
Gabral* Rkhd3 Inpp5f_v2 Ebf2
Rgs10 AK080843 Txnl2
MGI:2446326 BM117114 Mlstd2
Arrdc4 BB312372* Kcnq5
0710005119Rik BB264453* Dlgh2
BB182944 A1114950
BB077283 AA123443
AV328498 4933439C20Rik
4632427E13Rik
4632419K20Rik
2310047115Rik
Ht 2 1 1
Sh3gl3

For each combination of microarray-derived and assay-derived expression mode, the number of distinct genes that exhibited this combination is provided. Empty entries correspond to zero occurrences. The gene names are given except for genes that showed a biallelic mode of expression in both the microarray and assay experiments. New imprinted genes/transcripts are in bold. * indicates no SNP between Bb and Cast.

Table 4

Contingency table summarizing Supplementary Table S2 by contrasting the microarray-derived mode of expression (either M = maternal: uSLR ≥ .6, P = paternal: uSLR ≤ −.6 or B = biallelic) with the mode determined by the UpDp/UpD PCR and/or SNP sequencing assays

Mode of expression based on microarray measurement
Array-based Maternal Paternal Biallelic
expression mode: l M P B M P B M P B
Tissue Total
Pl 13 2 2 9
Ampd3 Dhcr7
Tspan4 Th
Em 28 9 2 4 13
A330103N21Rik AK080843 Ebf3
Ccndl BB264453 Gabra5
Ceacam11-14 Hbxap
Cebpa Rtn4
Pank3
Teadl
Br 30 1 5 8 11 5
Gabral* Rkhd3 Inpp5f_v2 Ebf2
Rgs10 AK080843 Txnl2
MGI:2446326 BM117114 Mlstd2
Arrdc4 BB312372* Kcnq5
0710005119Rik BB264453* Dlgh2
BB182944 A1114950
BB077283 AA123443
AV328498 4933439C20Rik
4632427E13Rik
4632419K20Rik
2310047115Rik
Ht 2 1 1
Sh3gl3
Mode of expression based on microarray measurement
Array-based Maternal Paternal Biallelic
expression mode: l M P B M P B M P B
Tissue Total
Pl 13 2 2 9
Ampd3 Dhcr7
Tspan4 Th
Em 28 9 2 4 13
A330103N21Rik AK080843 Ebf3
Ccndl BB264453 Gabra5
Ceacam11-14 Hbxap
Cebpa Rtn4
Pank3
Teadl
Br 30 1 5 8 11 5
Gabral* Rkhd3 Inpp5f_v2 Ebf2
Rgs10 AK080843 Txnl2
MGI:2446326 BM117114 Mlstd2
Arrdc4 BB312372* Kcnq5
0710005119Rik BB264453* Dlgh2
BB182944 A1114950
BB077283 AA123443
AV328498 4933439C20Rik
4632427E13Rik
4632419K20Rik
2310047115Rik
Ht 2 1 1
Sh3gl3

For each combination of microarray-derived and assay-derived expression mode, the number of distinct genes that exhibited this combination is provided. Empty entries correspond to zero occurrences. The gene names are given except for genes that showed a biallelic mode of expression in both the microarray and assay experiments. New imprinted genes/transcripts are in bold. * indicates no SNP between Bb and Cast.

The authors wish it to be known that, in their opinion, the first two authors should be regarded as joint First Authors

GEO accession no GSE4870

The authors thank Dr A. C. Ferguson-Smith for use of data on the UpD12 microarrays, C. V. Beechey for the T65H mouse translocation samples and L. A. Underkoffler and J. N. Collins for experimental assistance with some of the microarrays. The authors thank Dr B. S. Emanuel for human DNA/RNA samples. This work was supported by The Wellcome Trust (R.J.O. and T.R.M.), the BBSRC (R.J.O. and R.S.), EMBO (R.S.), The Guy's and St Thomas' Charity (R.J.O. and K.W.), The Generation Trust (A.J.W.) and Public Health Service Grant number GM58759 from the National Institutes of Health (R.J.O.). Funding to pay the Open Access publication charges for this article was provided by The Wellcome Trust.

Conflict of interest statement. None declared.

REFERENCES

1

McGrath, J. and Solter, D.

1984

Completion of mouse embryogenesis requires both the maternal and paternal genomes

Cell

37

179

–183

2

Surani, A., Barton, S.C., Norris, M.L.

1984

Development of reconstituted mouse eggs suggests imprinting of the genome during gametogenesis

Nature

308

548

–550

3

Morison, I.M., Ramsay, J.P., Spencer, H.G.

2005

A census of mammalian imprinting

Trends Genet

.

21

457

–465

4

Barlow, D.P.

1995

Gametic imprinting in mammals

Science

270

1610

–1613

6

Luedi, P.P., Hartemink, A.J., Jirtle, R.L.

2005

Genome-wide prediction of imprinted murine genes

Genome Res

.

15

875

–884

7

Nicholls, R.D. and Knepper, J.L.

2001

Genome organization, function, and imprinting in Prader–Willi and Angelman syndromes

Annu. Rev. Genomics Hum. Genet

.

2

153

–175

8

Monk, D. and Moore, G.E.

2004

Intrauterine growth restriction–genetic causes and consequences

Semin. Fetal Neonatal Med

.

9

371

–378

9

Feinberg, A.P.

2004

The epigenetics of cancer etiology

Semin. Cancer Biol

.

14

427

–432

10

Hochedlinger, K. and Jaenisch, R.

2003

Nuclear transplantation, embryonic stem cells, and the potential for cell therapy

N. Engl. J. Med

.

349

275

–286

11

Ogawa, H., Ono, Y., Shimozawa, N., Sotomaru, Y., Katsuzawa, Y., Hiura, H., Ito, M., Kono, T.

2003

Disruption of imprinting in cloned mouse fetuses from embryonic stem cells

Reproduction

126

549

–557

12

Simon, I., Tenzen, T., Reubinoff, B.E., Hillman, D., McCarrey, J.R., Cedar, H.

1999

Asynchronous replication of imprinted genes is established in the gametes and maintained during development

Nature

401

929

–932

13

Umlauf, D., Goto, Y., Cao, R., Cerqueira, F., Wagschal, A., Zhang, Y., Feil, R.

2004

Imprinting along the Kcnq1 domain on mouse chromosome 7 involves repressive histone methylation and recruitment of Polycomb group complexes

Nature Genet

.

36

1296

–1300

14

Lewis, A., Mitsuya, K., Umlauf, D., Smith, P., Dean, W., Walter, J., Higgins, M.J., Feil, R., Reik, W.

2004

Imprinting on distal chromosome 7 in the placenta involves repressive histone methylation independent of DNA methylation

Nature Genet

.

36

1291

–1295

15

Beechey, C.V., Ball, S.T., Townsend, K.M.S., Jones, J.

1997

The mouse chromosome 7 distal imprinting domain maps to G-bands F4/F5

Mamm. Genome

8

236

–240

16

Choi, J.D., Underkoffler, L.A., Collins, J.C., Marcheginani, S.M., Terry, N.A., Beechey, C.V., Oakey, R.J.

2001

Microarray expression profiling of tissues from mice with uniparental duplications of chromosomes 7 and 11 to identify imprinted genes

Mamm. Genome

12

758

–764

17

Oakey, R.J., Matteson, P.G., Litwin, S., Tilghman, S.M., Nussbaum, R.L.

1995

Nondisjunction rates and abnormal embryonic development in a mouse cross between heterozygotes carrying a (7,18) Robertsonian translocation chromosome

Genetics

141

667

–674

18

Georgiades, P., Watkins, M., Surani, M.A., Ferguson-Smith, A.C.

2000

Parental origin-specific developmental defects in mice with uniparental disomy for chromosome 12

Development

127

4719

–4728

19

Choi, J.D., Underkoffler, L.A., Collins, J.N., Williams, P.T., Golden, J.A., Loomes, K.M., Schuster, E.F., Jr, Wood, A.J., Oakey, R.J.

2005

A novel variant of Inpp5f is imprinted in brain and its expression is correlated with differential methylation of an internal exonic CpG island

Mol. Cell. Biol

.

25

001

–009

20

Smith, R.J., Arnaud, P., Kelsey, G.

2004

Identification and properties of imprinted genes and their control elements

Cytogenet. Genome Res

.

105

335

–345

21

Beechey, C.V.

1999

Imprinted genes and regions in mouse and human In Ohlsson, R. (Ed.).

Genomic Imprinting: An Interdisciplinary Approach, Results and Problems in Cell Differentialtion

Heidelberg, NY Springer-Verlag, Berlin pp.

303

–323

22

Cattanach, B.M. and Beechey, C.V.

1997

Genomic imprinting in the mouse: possible final analysis In Reik, W. and Surani, A. (Eds.).

Genomic Imprinting: Frontiers in Molecular Biology

Oxford, NY, Tokyo IRL Press Vol.

18

pp.

118

–145

23

Affymetrix.

2004

GeneChip® Expression Analysis Technical Manual

24

Hubbard, T.

2002

The Ensembl genome database project

Nucleic Acids Res

.

30

38

–41

25

Slater, G. and Birney, E.

2005

Automated generation of heuristics for biological sequence comparison

BMC Bioinformatics

6

31

26

Eyras, E., Caccamo, M., Curwen, V., Clamp, M.

2004

ESTGenes: alternative splicing from ESTs in Ensembl

Genome Res

.

14

976

–987

27

Wiehe, T., Gebauer-Jung, S., Mitchell-Olds, T., Guigó, R.

2001

SGP-1: prediction and validation of homologous genes based on sequence alignments

Genome Res

.

11

1157

–1183

28

Blanco, E., Parra, G., Guigó, R.

Using Geneid to Identify Genes

2003

NY John Wiley & Sons Inc In Current Protocols in bioinformatics, Vol 1, Baxevamis,A and Davison,D. (eds)

29

Burge, C. and Karlin, S.

1997

Prediction of complete gene structures in human genomic DNA

J. Mol. Biol

.

268

78

–94

30

Gough, J., Karplus, K., Hughey, R., Chothia, C.

2001

Assignment of homology to genome sequences using a library of hidden markov models that represent all proteins of known structure

J. Mol. Biol

.

313

903

–919

31

Korf, I., Flicek, P., Duan, D., Brent, M.R.

2001

Integrating genomic homology into gene structure prediction

Bioinformatics

1

S1

–S9

32

Stanke, M. and Waack, S.

2003

Gene prediction with a hidden-markov model and a new intron submodel

Bioinformatics

19

ii225

–ii225

33

Kent, J.W.

2002

BLAT—the BLAST-like alignment tool

Genome Res

.

12

656

–664

34

Affymetrix.

2002

Statistical Algorithms Description Document

35

Irizarry, R., Hobbs, B., Collin, F., Beazer-Barclay, Y., Antonellis, K., Scherf, U., Speed, T.

2003

Exploration, normalization, and summaries of high density oligonucleotide array probe level data

Biostatistics

4

249

–264

36

Takada, S., Tevendale, M., Baker, J., Goergiades, P., Campbell, E., Freeman, T., Johnson, M.H., Paulson, M., Ferguson-Smith, A.C.

2000

Delta-like and Gtl2 are reciprocally expressed, differentially methylated linked imprinted genes on mouse chromosome 12

Curr. Biol

.

10

1135

–1138

37

Allison, D.B., Cui, X., Page, G.P., Sabripour, M.

2006

Microarray data analysis: from disarray to consolidation and consensus

Nature Rev.Genet

.

7

55

–65

38

Piras, G., El Kharroubi, A., Kozlov, S., Escalante-alcalde, D., Hernandez, L., Copeland, N.G., Gilbert, D.J., Jenkins, N.A., Stewart, C.L.

2000

Zac1 (Lot1) , a potential tumor supressor gene, and the gene for ε-Sarcoglycan are maternally imprinted genes: identification by a subtractive screen of novel uniparental fibroblast lines

Mol. Cell. Biol

.

20

3308

–3315

39

Mizuno, Y., Sotomaru, Y., Katsuzawa, Y., Kono, T., Meguro, M., Oshimuru, M., Kawai, J., Tomaru, Y., Kiyosawa, H., Nikaido, I., et al.

2002

Asb4, Ata3 and Dcn are novel imprinted genes identified by high- throughput screening using RIKEN cDNA microarray

Biochem. Biophys. Res. Commun

.

290

1499

–1505

40

Fitzpatrick, G.V., Soloway, P.D., Higgins, M.J.

2002

Regional loss of imprinting and growth deficiency in mice with a targeted deletion of KvDMR1

Nature Genet

.

32

426

–431

41

Shi, W., Krella, A., Orth, A., Yu, Y., Fundele, R.

2005

Widespread disruption of genomic imprinting in adult interspecies mouse (Mus) hybrids

Genesis

43

99

–107

42

Buettner, V.L., Walker, A.M., Singer-Sam, J.

2005

Novel paternally expressed intergenic transcripts at the mouse Prader–Willi/Angelman Syndrome locus

Mamm. Genome

16

219

–227

43

Landers, M., Bancescu, D.L., Le Meur, E., Rougeulle, C., Glatt-Deeley, H., Brannan, C., Muscatelli, F., Lalande, M.

2004

Regulation of the large (∼1000 kb) imprinted murine Ube3A antisense transcript by alternative exons upstream of Snurf/Snrpn

Nucleic Acids Res

.

32

3480

–3492

44

Le Meur, E., Watrin, F., Landers, M., Sturny, R., Lalande, M., Muscatelli, F.

2005

Dynamic developmental regulation of the large non-coding RNA associated with the mouse 7C imprinted chromosomal region

Dev. Biol

.

286

587

–600

45

Monk, D., Arnaud, P., Apostolidou, S., Hills, F.A., Kelsey, G., Stanier, P., Feil, R., Moore, G.E.

2006

Limited evolutionary conservation of imprinting in the human placenta

Proc. Natl Acad. Sci. USA

103

6623

–6628

46

Nikaido, I., Saito, C., Mizuno, Y., Meguro, M., Bono, H., Kadomura, M., Kono, T., Morris, G.A., Lyons, P.A., Oshimura, M., et al.

2003

Discovery of imprinted transcripts in the mouse transcriptome using large-scale expression profiling

Genome Res

.

13

1402

–1409

47

Ruf, N., Dunzinger, U., Brinckmann, A., Haaf, T., Nurnberg, P., Zechner, U.

2006

Expression profiling of uniparental mouse embryos is inefficient in identifying novel imprinted genes

Genomics

Vol

87

4

509

–519

48

Paulsen, M., Davies, K., Hu, R.-J., Feinberg, A.P., Maher, E.R., Reik, W., Walter, J.

1998

Syntenic organization of the mouse distal chromosome 7 imprinting cluster and the Beckwith–Wiedemann syndrome region in chromosome 11p15.5

Hum. Mol. Genet

.

7

1149

–1159

49

Engemann, S., Strodicke, M., Paulsen, M., Franck, O., Reinhardt, R., Lane, N., Reik, W., Walter, J.

2000

Sequence and functional comparison in the Beckwith–Wiedemann region: implications for a novel imprinting centre and extended imprinting

Hum. Mol. Genet

.

9

2691

–2706

50

Higashimoto, K., Soejima, H., Yatsuki, H., Joh, K., Uchiyama, M., Obata, Y., Ono, R., Wang, Y., Xin, Z., Zhu, X., et al.

2002

Characterization and imprinting status of OBPH1/Obph1 gene: implications for an extended imprinting domain in human and mouse

Genomics

80

575

–584

51

Reik, W., Brown, K.W., Schneid, H., Le Bouc, Y., Bickmore, W., Maher, E.R.

1995

Imprinting mutations in the Beckwith–Wiedemann syndrome suggested by altered imprinting pattern in the IGF2-H19 domain

Hum. Mol. Genet

.

4

2379

–2385

52

Thorvaldsen, J., Duran, J.L., Bartolomei, M.S.

1998

Deletion of the H19 differentially methylated domain results in loss of imprinted expression of H19 and Igf2

Genes Dev

.

12

3693

–3702

53

Smilinich, N.J., Day, C.D., Fitzpatrick, G.V., Caldwell, G.M., Lossie, A.C., Cooper, P.R., Smallwood, A.C., Joyce, J.A., Schofield, P.N., Reik, W., et al.

1999

A maternally methylated CpG island in KvLQT1 is associated with an antisense paternal transcript and loss of imprinting in Beckwith–Wiedemann syndrome

Proc. Natl Acad. Sci. USA

96

8064

–8069

54

Moore, T., Constancia, M., Zubair, M., Bailleul, B., Feil, R., Sasaki, H., Reik, W.

1997

Multiple imprinted sense and antisense transcripts, differential methylation and tandem repeats in a putative imprinting control region upstream of mouse Igf2

Proc. Natl Acad. Sci. USA

94

12509

–12514

55

Constancia, M., Hemberger, M., Hughes, J., Dean, W., Ferguson-Smith, A., Fundele, R., Stewart, F., Kelsey, G., Fowden, A., Sibley, C., et al.

2002

Placental-specific IGF-II is a major modulator of placental and fetal growth

Nature

417

945

–948

56

Gerard, M., Hernandez, L., Wevrick, R., Stewart, C.L.

1999

Disruption of the mouse necdin gene results in early post-natal lethality

Nature Genet

.

23

1999

–1202

© 2006 The Author(s) This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License ( http://creativecommons.org/licenses/by-nc/2.0/uk/ ) which permits unrestricted non-commerical use, distribution, and reproduction in any medium, provided the original work is properly cited.

I agree to the terms and conditions. You must accept the terms and conditions.

Submit a comment

Name

Affiliations

Comment title

Comment

You have entered an invalid code

Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.

Citations

Views

Altmetric

Metrics

Total Views 968

651 Pageviews

317 PDF Downloads

Since 2/1/2017

Month: Total Views:
February 2017 8
March 2017 4
April 2017 1
May 2017 9
June 2017 4
July 2017 2
August 2017 4
September 2017 2
October 2017 2
November 2017 1
December 2017 7
January 2018 5
February 2018 10
March 2018 20
April 2018 11
May 2018 7
June 2018 6
July 2018 13
August 2018 4
September 2018 7
October 2018 10
November 2018 18
December 2018 8
January 2019 12
February 2019 7
March 2019 14
April 2019 14
May 2019 19
June 2019 9
July 2019 7
August 2019 10
September 2019 6
October 2019 8
November 2019 22
December 2019 16
January 2020 16
February 2020 31
March 2020 13
April 2020 11
May 2020 8
June 2020 11
July 2020 12
August 2020 12
September 2020 7
October 2020 20
November 2020 15
December 2020 3
January 2021 5
February 2021 2
March 2021 8
April 2021 6
May 2021 1
June 2021 6
July 2021 13
August 2021 3
September 2021 7
October 2021 6
November 2021 8
December 2021 5
January 2022 14
February 2022 5
March 2022 4
April 2022 12
May 2022 8
June 2022 9
July 2022 9
August 2022 9
September 2022 12
October 2022 15
November 2022 10
December 2022 7
January 2023 9
February 2023 5
March 2023 2
April 2023 3
May 2023 7
June 2023 3
July 2023 6
August 2023 11
September 2023 16
October 2023 7
November 2023 23
December 2023 57
January 2024 20
February 2024 17
March 2024 17
April 2024 19
May 2024 17
June 2024 11
July 2024 16
August 2024 14
September 2024 9
October 2024 29

Citations

56 Web of Science

×

Email alerts

Citing articles via

More from Oxford Academic