Abstract
Sex differences in gene expression are widespread in the liver, where many autosomal factors act in tandem with growth hormone signaling to regulate individual variability of sex differences in liver metabolism and disease. Here, we compare hepatic transcriptomic and epigenetic profiles of mouse strains C57BL/6J and CAST/EiJ, representing two subspecies separated by 0.5–1 million years of evolution, to elucidate the actions of genetic factors regulating liver sex differences. We identify 144 protein coding genes and 78 lncRNAs showing strain-conserved sex bias; many have gene ontologies relevant to liver function, are more highly liver-specific and show greater sex bias, and are more proximally regulated than genes whose sex bias is strain-dependent. The strain-conserved genes include key growth hormone-dependent transcriptional regulators of liver sex bias; however, three other transcription factors, Trim24, Tox, and Zfp809, lose their sex-biased expression in CAST/EiJ mouse liver. To elucidate the observed strain specificities in expression, we characterized the strain-dependence of sex-biased chromatin opening and enhancer marks at cis regulatory elements (CREs) within expression quantitative trait loci (eQTL) regulating liver sex-biased genes. Strikingly, 208 of 286 eQTLs with strain-specific, sex-differential effects on expression were associated with a complete gain, loss, or reversal of the sex differences in expression between strains. Moreover, 166 of the 286 eQTLs were linked to the strain-dependent gain or loss of localized sex-biased CREs. Remarkably, a subset of these CREs apparently lacked strain-specific genetic variants yet showed coordinated, strain-dependent sex-biased epigenetic regulation. Thus, we directly link hundreds of strain-specific genetic variants to the high variability in CRE activity and expression of sex-biased genes and uncover underlying genetically-determined epigenetic states controlling liver sex bias in genetically diverse mouse populations.
Author summary
Male-female differences in liver gene expression confer sex differences in diverse biological processes relevant to human health and disease but are difficult to model in inbred mice given their identical genetic backgrounds. Outbred mice provide some variability, but cross-strain studies of sex bias in rodents have not been well studied. Here we elucidate the actions of genetic factors regulating liver sex differences in two Diversity Outbred mouse founder mouse strains, C57BL/6 and CAST/EiJ. We find that many of the strain differences in sex-biased gene expression can be linked to the gain or loss of a cis regulatory element associated with one or more strain-specific sequence variants. Strikingly, in many cases, the associated cis regulatory element lacked strain-specific variants, yet was subject to coordinated, strain-dependent epigenetic regulation. Thus, harnessing the power of naturally occurring genetic diversity of Diversity Outbred mice, we integrated biological data at the genetic, epigenetic, and transcriptomic levels across evolutionary divergent mouse strains to discover hundreds of localized genomic regions that control phenotypic sex differences in the liver. These findings may serve as a model for studies of human genetic variation and the effect of population-wide variation on sex differences in health and disease.
Introduction
Many vertebrate tissues, including non-reproductive tissues, show significant sex differences in their gene expression profiles, metabolic and physiological properties, and patterns of disease susceptibility [1–3]. Underlying regulatory mechanisms are best studied in the liver, where there is extensive transcriptomic and regulatory sex bias in fish [4,5], rats [6,7], mice [8–10] and humans [11,12]. In mouse liver, hundreds of genes are expressed in a sex-dependent manner, including protein-coding genes [13], miRNAs [14,15] and lncRNA genes [16–18]. Phenotypically, these sex differences in expression contribute to sex differences in chemical sensing and metabolism [19–21], response to injury [22,23], and susceptibility to disease [24–27].
Growth hormone (GH) has been directly implicated as the major proximal regulator of sexual dimorphism in the liver. Pituitary GH secretion patterns, which are controlled by the hypothalamus in a sex-dependent manner [28,29], activate GH signaling to the nucleus through the JAK/STAT pathway [30,31] and are crucial to hepatic sex differences in both rodents and humans [20,32–34]. Thus, sex-biased gene expression in the liver is substantially lost when GH secretion is ablated (>90% of sex-biased genes) [35,36] or following continuous GH infusion, which overrides the endogenous male, pulsatile plasma GH profile (74% of sex-biased genes) [13]. Major dysregulation of hepatic sex-biased genes also occurs upon loss of the GH receptor-activated proximal transcriptional regulator STAT5 [9,37,38] or its downstream targets, notably the transcriptional repressors Bcl6 [39,40] and Cux2 [41].
Genome evolution results in species differences in transcription factor binding at cis regulatory elements (CREs), with a strong preference for evolution of regulatory sequences at enhancers as compared to gene promoters [42,43]. Analysis of such evolutionary changes at CREs between closely related species, or across strains within a species, provides an opportunity to discover functional roles of specific regulatory elements [44–46]. Genetic diversity across mouse strains and subspecies results in well-characterized phenotypic [47,48], transcriptomic [49–51], and epigenetic differences in the liver [44–46]. Furthermore, extensive transcriptomic and epigenetic studies on liver sex differences in the outbred mouse strain CD-1 have identified large numbers of GH-regulated CREs that differ between the sexes [38,52–54]. The sex bias of hepatic gene expression has also been described in several inbred mouse strains [8,10,55]. Underlying mechanisms for genetically-based differences between strains are poorly understood [49,56–59], primarily because most studies of sex bias in mouse liver have focused on strains of primarily M. m. domesticus genetic background (i.e., CD-1 and C57BL/6J) and did not include wild-derived strains.
The Diversity Outbred (DO) mouse model, where known genetic variants from eight well-characterized inbred founder mouse strains have been reshuffled through a specific multi-generation breeding program [60,61], is an important resource for studying the contributions of genetic variation to diversity. The genetic diversity of individual DO mice [62] is comparable to the diversity of the global human population [63] and serves as a relevant model for diversity in gene expression and disease in humans. Livers from individual DO mice differ in their responses to chemical exposure [64] and western diet [65], and show marked differences in expression of sex-biased genes [17]. Furthermore, recent work from this laboratory analyzing RNA-seq datasets from 438 individual DO mice identified 1,137 expression quantitative trait loci (eQTLs), each harboring genetic variants with a significant impact on the expression of sex-biased genes [49]. However, the loci identified are quite large (median width = 1.84 Mb) and contain large numbers of strain-specific SNPs and Indels, making it difficult to link specific genetic variants to the variable expression of individual sex-biased genes.
Here, we use a combination of DNase-seq, ChIP-seq and RNA-seq to capture sex differences, and their strain dependence, at the epigenetic and transcriptomic levels for two DO mouse founder strains, C57BL/6J (B6) and CAST/EiJ (CAST) mice. These strains—B6, an inbred laboratory mouse strain composed of primarily subspecies Mus musculus domesticus genetic background, and the wild-derived CAST strain, of subspecies Mus musculus castaneus—comprise two of the eight founder strains originally used to generate DO mice [60,61] and are separated by 0.5–1 million years of evolution [66,67]. We identify > 200 strain-conserved sex-biased protein coding and lncRNA genes between B6 and CAST mouse liver, including several core transcription factors regulating liver sex differences, which likely underpin the conservation of phenotypic sex bias across strains. Sex-biased expression was lost in CAST liver for three B6 sex-biased transcriptional regulators, which may contribute to the strain-dependence of sex-biased gene expression through a trans effect. Further, we identify hundreds of strain-conserved, sex-biased CREs, 80% of which have properties of gene-distal enhancers, and we elucidate the strain-specific gain or loss of many hundreds of other sex-biased CREs associated with genetic variation within eQTL regions. Importantly, a majority of the strain differences that we evaluated in the set of eQTLs regulating strain-specific, sex-biased gene expression were directly associated with a gain or loss of CREs, but remarkably, a subset of these CREs were devoid of strain-specific SNPs/Indels. Thus, by integrating biological data at the genetic, epigenetic, and transcriptomic levels across evolutionarily divergent inbred mouse strains, we identify genomic sequences likely to serve as causal regulators of the transcriptional activity of a significant subset of the sex-biased murine liver transcriptome.
Results
Strain-shared and strain-unique sex-biased genes in B6 and CAST mouse liver
We performed RNA-seq to identify genes showing sex-biased expression in B6 and CAST mouse livers. Results were compared to prior data for sex-biased genes in livers of the outbred mouse strain CD-1 [13], which is most similar to the DO founder mouse strain NOD/ShiLt and is particularly useful due to its inherent genetic variability [68]. 946 protein-coding genes and 751 lncRNA genes showed significant liver sex-bias in one or more strains (Fig 1A and S1 Table). We observed a greater overlap of sex-biased genes between CD-1 and B6 relative to CAST (229 protein coding genes versus 144, respectively; Fig 1A), which is likely because both strains are composed of primarily subspecies Mus musculus domesticus genetic background [66–68]. Comparing the two evolutionarily distant inbred strains, B6 and CAST, we identified 144 protein-coding genes and 78 lncRNA genes with strain-shared sex bias (defined as significant sex bias in both B6 and CAST). These strain-shared sex-biased genes were clustered and annotated based on their relative expression across individual livers from each strain and sex (Fig 1B). Importantly, the directionality of sex bias was conserved between strains for almost all the strain-shared sex biased genes (217 of 222 genes; Fig 1B and 1C; examples in S1 Fig and full gene lists in Sheets A and B in S1 Table). However, only 29% of the core strain-shared sex-biased genes (43/144 protein-coding genes, 21/78 lncRNAs) showed the same expression pattern in livers of both strains (clusters MA and FA; Fig 1B and 1C). The remaining 71% of strain-shared genes showed strain differences in the absolute expression level (FPKM) and magnitude of sex bias (e.g., genes in clusters MB, FB1, and FB2 showed higher expression in B6 than CAST). Accordingly, while most of the variation in expression of strain-shared sex-biased genes is associated with differences in sex (PC1, 60–66% of variance), substantial variation is explained by strain differences (PC2, 22–26% of variance; S1E and S1F Fig). Finally, we identified genes whose sex-biased expression in liver is specific to one strain. Thus, B6-unique: 301 protein-coding genes, 289 lncRNAs; and CAST-unique: 207 protein-coding genes, 187 lncRNAs. A majority of these strain-unique sex-biased genes are more highly expressed in the strain showing significant sex-bias (S2 and S3 Figs and Sheets C-F in S1 Table).
Functional roles of strain-shared vs strain-unique sex-biased genes
Despite substantial evolutionary distance between B6 and CAST mice, sex-biased expression is maintained for a core set of functionally relevant and liver-specific genes. The core set of 144 strain-shared sex-biased protein-coding genes (Fig 1B) includes many Cyp, Mup, and Sult genes, and showed strong enrichment for molecular function gene ontologies relevant to liver function, including monooxygenase activity and small molecule binding/lipocalin (Fig 1D). In contrast, only one Gene Ontology term (transporter activity) was significantly enriched in the set of 301 B6-unique sex-biased protein-coding genes, and no molecular function terms were enriched in the 207 CAST-specific gene set (FDR<0.01; S2 Table). Furthermore, the strain-shared sex-biased genes are expressed in a highly liver-specific manner, as compared to either the strain-unique gene sets or all liver expressed genes, based on comparisons to 21 mouse ENCODE tissues [69] (Mann-Whitney (M-W) p < 0.001; Fig 1E).
The major GH-regulated or GH-responsive transcription factors (TFs) implicated in liver sex differences in CD-1 mice were expressed at similar levels in both mouse strains, consistent with a model of evolutionary conservation of functionally relevant genes. These include the GH-responsive TFs STAT5b [9], Foxa2 [70] and Hnf4a [37], and the GH-regulated, sex-biased TFs Cux2 [71], Bcl6 [40] and Sall1 [13] (S4A and S4B Fig and Sheet A in S3 Table). The sex-biased expression of five other TF genes was strain-dependent (Sheets B and C in S3 Table); these include three B6-unique sex-biased TFs previously identified as being GH-responsive (Trim24, Tox, and Zfp809; S4C Fig) [13,71,72] and two CAST-unique sex-biased TFs with no known roles in liver sex differences (Id3, Rnf141; S4D Fig). Conceivably, the three B6 mouse-specific TFs may contribute to the larger number of sex-biased genes found in B6 (and CD-1) mouse liver compared to CAST mouse liver (Figs 1A and S4E).
Strain-shared and strain-unique sex-biased cis regulatory elements
We sought to identify regulatory elements for sex-biased liver gene expression, both those that are common and those that differ between B6 and CAST mice. We sequenced genome-wide ChIP-seq libraries for H3K27ac, which marks active enhancers and promoters, and we performed DNase-seq to identify open chromatin regions in liver nuclei collected from B6 and CAST mice of both sexes (Sheets B-D in S4 Table). These analyses identified thousands of regulatory elements (H3K27ac peaks and DNase hypersensitive sites (DHS)) in both strains and sexes (S5 Table). Using ChIP-qPCR and DNase-qPCR primers anchored at known sex-biased enhancers nearby strain-dependent sex-biased genes, we verified the presence of both B6-specific and CAST-specific epigenetic marks (S5 Fig). Furthermore, by using diffReps [73] to compare data for male vs female livers within each strain, followed by filtering to increase robustness (see Methods), we discovered ~2,000 robust sex-biased H3K27ac peak regions, and ~1,000 robust sex-biased DHS in livers of each strain (S6A–S6D Fig), with consistent sex bias seen across biological replicates (S6E Fig). A subset of these sex-biased regulatory regions was shared between strains and showed a concordant sex-bias, while many others were unique to one strain (Fig 2A and 2B). Thus, of 2,288 robust sex-biased B6 liver H3K27ac peak regions, 441 were shared with CAST mice, of which 98% showed concordant sex-bias (431 of 441; 198 female-biased, 233 male-biased) (Sheet E in S5 Table). Similarly, 92 sex-biased open chromatin regions (DHS) were shared between B6 and CAST mice, with 97% showing concordant sex-bias (89 of 92; Sheet D in S5 Table). Thus, many sex-biased regulatory regions maintain consistent sex-bias, despite the evolutionary divergence between B6 and CAST mice.
We use the term cis regulatory elements (CREs) to describe both strain-conserved and strain-unique regions with the potential to regulate genes in cis, i.e., intra-chromosomally or within the same Topologically Associating Domain (TAD) [74]. Overall, ~80% of the regulatory elements we identified—both sex-biased and sex-independent sequences—are outside of promoter regions (>2.5 kb), i.e., are enhancer regions (S6F Fig). Globally, strain-shared H3K27ac peaks and DHS regions showed greater sequence conservation across vertebrates than strain-unique peaks and regions (S6G Fig); however, the strain-shared sex-biased H3K27ac peaks and DHS regions did not show greater sequence conservation than the corresponding strain-unique regions. Thus, global sequence conservation alone does not explain the gain or loss of sex bias between strains.
Two examples of strain-shared sex-biased CREs neighboring core sex-biased genes are shown in Fig 2C: the male-biased complement component gene C6 (16.4-fold average male bias across B6, CAST and CD-1 livers) and the female-biased nucleotidase Nt5e (5.7-fold average female bias). Extensive sex-bias, both locally at the promoter and at more distal enhancers, is seen for H3K27ac mark accumulation and chromatin opening with a directionality that matches the sex bias of gene expression. Other examples of core, strain-shared male-biased genes (Gstp1, Elovl3) and female-biased genes (Acot3, Acot4, Cyp4a14) showed uniform directionality in sex-biased enhancer activity and gene expression across strains (S7 and S8 Figs).
In closely related mammals, gene expression shows greater conservation than TF-binding or chromatin accessibility [44]. Consistent with this, a smaller fraction of sex-biased CREs was conserved across strains as compared to the fraction of genes whose sex-biased expression is conserved across strains. Thus, while 26–33% of sex-biased genes are shared between B6 and CAST liver (Fig 1A), only 17–19% of sex-biased H3K27ac peak regions and 7–9% of sex-biased DHS are strain-shared (Fig 2A and 2B). This result can, in part, be explained by the complexity of enhancer coordination, where a conserved pattern of transcriptional output may emerge from the integration of the regulatory activity of multiple enhancers showing variability with regards to sex-bias of their activity. Supporting this idea, we observed both strain-shared and strain-unique sex-bias at several CREs nearby strain-shared sex-biased genes, such as the male-biased gene Nudt7 and the female-biased gene Cux2 (S7C and S8C Figs), which display a similar magnitude of sex bias between strains.
Sex-biased CREs are closer to strain-shared than strain-unique sex-biased genes
Strain-shared sex-biased genes are generally closer than strain-unique sex-biased genes to CREs with matched sex bias. For example, 23.6% of all strain-shared sex-biased genes have a strain shared sex-biased CRE within 2 kb of their TSS, while only 15.2% of B6-unique sex-biased genes have a B6-unique sex-biased CRE within 2 kb of their TSS (Fig 3A vs. Fig 3B). The frequency of a strain-shared sex-biased promoter CRE is even higher (39%) for the 43 strain-shared sex-biased genes that show a consistent expression level in both strains (Fig 3A; genes in clusters FA or MA of Fig 1B). Both the strain-shared (Fig 3A) and the strain-unique sex-biased gene sets are more distant from sex-biased CREs of the opposite sex (Fig 3B and 3C; KS p < 0.0001). The strain-unique sex-biased genes were also significantly closer to strain-specific CREs in the same strain than similarly sex-biased CREs in the other strain (Fig 3B and 3C). After grouping sex-biased genes based on distance to correspondingly sex-biased CREs (proximal CRE, < 20 kb; distal CRE, > 20 kb but within the same TAD), we found that core, strain-shared sex-biased genes are more likely than strain-unique genes to be associated with a correspondingly sex-biased promoter region CRE (Fig 3D).
In a prior study, genes with strong liver sex bias were often subject to proximal regulation [52]. Consistent with this, strain-conserved sex-biased genes, with their greater frequency of proximal sex-biased CREs, showed a greater magnitude of sex-bias than strain-unique sex-biased genes (Fig 3E). However, the 43 strain-shared sex-biased genes with a strain-conserved expression pattern were not significantly more sex-biased than other 101 core sex-biased genes (M-W p = 0.698). Thus, the greater strength of sex bias for the strain-conserved genes cannot entirely be explained by their closer proximity to CREs.
Enrichment of strain-specific genetic variants at sex-biased CREs
To investigate the impact of genetic variants on sex-biased gene expression, we compared the above sets of strain-specific CREs to a set of 1,172 eQTL regions implicated in the variation of sex-biased gene expression across a set of 438 male and female DO mouse livers [49]. Each eQTL region can be linked, with directionality of the eQTL’s effects on sex-biased gene expression, to specific genetic variants originating from one or more of the eight DO mouse inbred founder strains. Two of the eight strains, CAST and B6, are the regulating strain (see Methods for definition) for 491 of the 1,172 eQTLs, which impact the expression of 481 sex-biased genes (Sheets A and B in S6 Table). Many more eQTLs and genetic variants are specific to CAST mice (406 eQTLs) than B6 mice (85 eQTLs) (S9A Fig), which is the only m. castaneus subspecies among the 8 DO mouse founder strains. The vast majority of eQTLs are cis to the affected gene: 78% (n = 383 eQTLs) mapped to the same TAD as the regulated gene and 14% (n = 67 eQTLs) were outside of the TAD but mapped to the same chromosome vs. only 8% (n = 41 eQTLs) mapped to a different chromosome (Fig 4A). In addition to relying on our prior characterization of eQTLs related to sex bias [49], we reanalyzed the underlying DO mouse datasets by adding a sex-eQTL interaction term into the model [75]. This analysis revealed that 168 of the 491 eQTLs with CAST or B6 as the regulating strain show a significant difference in LOD score between male and female liver (Sheet A in S6 Table). The genes regulated by these 168 eQTLs tend to exhibit greater sex bias than the genes regulated by the full set of 491 eQTLs (3.85 vs 2.25 median absolute fold-change; p = 6.12e-27), while the maximum expression was not significantly higher (p = 0.0831; S9B Fig). The subset comprised of 168 eQTLs was designated robust sex-specific eQTLs, while those only identified in the prior analysis, where each sex was treated separately [49], were designated lenient eQTLs.
One limitation of eQTL analysis is its comparatively low resolution (median interval = 1.7 Mb for 491 eQTLs for sex-biased genes) (S9C and S9F Fig). Consequently, the above set of 491 eQTLs impacting sex-biased genes encompassed a full 34% of all 10.2 million genome-wide SNPs/Indels between B6 and CAST mice (S9D Fig). Moreover, genetic variants differentiating CAST and B6 mice occur every ~100 bp [46] (every ~275 bp when considering variants unique to CAST or B6 mice relative to the six other DO founder strains; see Methods) and often occur within large, megabase-scale haplotype blocks, each containing thousands of SNPs and Indels. We therefore sought to significantly narrow the genomic region for consideration within each eQTL region. First, we limited our analysis to variants within the genomic region defined by the intersection of the eQTL region and the TAD containing the TSS of the regulated gene (intra-TAD interval, S9E Fig). This reduced the median interval size 4-fold, to 0.42 Mb, and decreased the number of genetic variants per eQTL region 3-fold, from 4,875 to 1,578 SNPs/Indels (S9F Fig). Second, we only considered as causative SNP/Indel candidates those that map to sex-biased CREs within the intra-TAD interval (c.f., [76]). This further reduced the median number of variants per eQTL by ~30-fold, from 1,578 to 51 (S9F Fig). We validated this approach by comparing the enrichment of strain-specific genetic variants at CREs within the intra-TAD interval of each eQTL to that of the full length eQTL (Fig 4B). Thus, we observed weak enrichment of strain-specific variants at sex-biased CREs when considering the full eQTL regions, but found a highly significant, 2.5-fold enrichment within intra-TAD intervals (p<0.001 for intra-TAD interval variants vs p = 0.278 genome-wide). In an important control, the enrichment in intra-TAD intervals was marginal when strain-specific variants at all CREs were considered. Furthermore, no enrichment of strain-specific variants at sex-biased CREs was found when we considered all CAST-specific or all B6-specific genetic variants genome-wide (1.01 observed/expected) (Fig 4B, first purple bar). Finally, variants were rarely found within a TF-binding motif (Fig 4B, green bars), consistent with prior reports [46,77,78].
Strong genetic effects of DO mouse liver eQTLs reflect the gain or loss of cis regulation
286 of the above full set of 491 eQTLs regulating genes that exhibit sex-biased expression in B6 or CAST mouse liver can be categorized based on the mechanism whereby the eQTL either reduces or enhances sex differences in expression, as previously described [49] (S9G and S9H Fig). Specifically, eQTLs in categories #1–4 impart either a reduction in, or a total loss of, sex bias in the regulating mouse strain, while eQTLs in categories #5–8 confer either an increase in, or an outright gain of, sex-specificity in the regulating strain (Fig 4C) [49] (see Methods for definitions). These categories were further subdivided based on the sex in which the eQTL has a higher LOD score and the directionality of effect (e.g., Category #1 eQTLs result in repression of a male-biased gene in the regulating strain). Remarkably, 208 of the 286 categorized eQTLs (73%) resulted in either a loss, a gain, or a reversal of sex bias when comparing B6 and CAST mouse liver (Fig 4C, right). Sex bias is retained but is either reduced (eQTL categories #1–4) or is increased (eQTL categories #5–8) in magnitude in the regulating strain for the other 78 eQTLs. The percent of eQTLs resulting in a gain or a loss of sex bias was as high as 91% for category #1 eQTLs (repression of male-biased genes in male liver) (Fig 4D). Mechanistically, 166 of the 286 eQTLs can potentially be explained by a strain-specific sex-bias in one or more CREs that harbor strain-specific genetic variants (Fig 4E and Sheet G in S6 Table). For eQTL categories #1–4, this is indicated by the absence in the regulating strain of one or more sex-biased CREs, which, in the other, non-regulating strain, show the same sex bias as the sex-biased gene. For eQTL categories #5–8, this is indicated by the presence in the regulating strain, but not in the non-regulating strain, of one or more sex-biased CREs showing the same sex bias as the regulated gene. Together, these findings, although correlative in nature, lend strong support for the proposal that the genetic factors identified by these 166 eQTLs impart the observed gain or loss of sex-biased gene expression primarily through the gain or loss of one or more of the associated sex-biased CREs. Specific examples, all robust sex-biased eQTLs, are presented for each eQTL category in Figs 5–8, below.
eQTL-gene relationships where a single, potentially causal, sex-biased CRE is gained or lost
For 61 of the above 166 eQTLs, we identified only one sex-biased CRE that contains genetic variant(s) in the regulating strain, and which could thus be responsible for the observed gain or loss of sex bias (Sheets A and G in S6 Table). Examples include the male-biased genes Cspg5 and Olfm2 (Figs 5A and S10A) and the female-biased genes Ptger3 and Enpp1 (Figs 5B and S10B). Cspg5 expression is 20-fold lower in CAST male liver, as compared to B6 male liver (category #1 eQTL), and only a single male-biased CRE, within the 5’ UTR of Cspg5, contains CAST-specific genetic variants. Although other variants that are not CAST-specific are present in the region, all but two are outside of the indicated promoter-proximal CRE (Fig 5A; Unique versus All variants). The SNP within this CRE (G to C in CAST mice) is associated with the loss of male-biased H3K27ac marks at the Cspg5 promoter in CAST liver and loss of the 20-fold male bias in expression seen in B6 liver (Fig 5A). Conversely, the strong up regulation of the female-biased gene Ptger3 in CAST female compared to B6 female liver (category #8 eQTL) is associated with a single female-biased CRE at the TSS and 5’ UTR of Ptger3 (Fig 5B). The three CAST-specific SNPs in this CRE are associated with a gain of female-specific H3K27ac marks and with up regulation of Ptger3 expression in CAST female liver (5.7-fold female-bias in CAST liver vs no sex bias in B6 liver; Fig 5B). For both Cspg5 and Ptger3, the sex-biased CREs shown are the only candidate regulators harboring strain-specific SNPs within the eQTL region (Sheet A in S6 Table).
Enhancement of the sex-bias of expression can also be associated with loss of positive regulatory elements (eQTL categories #6 and #7). Thus, the female-biased expression of Slc16a5 increases from 5.5-fold in B6 mouse liver to 48.7-fold in CAST liver, primarily due to a decrease in basal expression in CAST males (category #6 eQTL; Fig 6A). Slc16a5 expression, DHS, and H3K27ac marks at the Slc16a5 promoter are equivalent in B6 and CAST female liver, however, all three are reduced in CAST male liver compared to B6 male liver in association with CAST sequence variants (Fig 6B). Conversely, the repression of the male-biased gene Bok seen in female CAST liver (coefficient -0.91) but not in male CAST liver results in an increase in male bias (S10C Fig). This repression in female CAST liver can be explained by the decrease in H3K27ac marks within the Bok gene body in female CAST liver compared to female B6 liver (S10D Fig), and consequently, there is a female-biased CRE in CAST but not B6 liver (category #7 eQTL; S10C Fig). These examples illustrate how genetic variants within a single CRE may lead to a gain or loss of sex-biased expression.
Coordinated gain or loss of sex bias across multiple CREs within an eQTL
A majority of the eQTLs in each category show a gain or loss of multiple sex-biased CREs (Fig 4E; 105 of the 166 sex-biased eQTLs discussed above), and hence their regulation and the genetic mechanisms governing the strain-dependence of their sex-biased expression is intrinsically more complex than the cases of single sex-biased CREs discussed above. Indeed, as illustrated by the examples below, CREs within these eQTLs typically showed coordinated, strain-dependent differences in their sex-biased enhancer activity, which in many cases cannot be explained by a strain-specific SNP or Indel within the CRE.
Moxd1 shows a 12-fold increase in male bias in CAST compared to B6 liver (from 64-fold to 776-fold), due to a large increase in expression in CAST male liver (Fig 7A, category #5 eQTL). This increase is associated with increased activity of multiple male-biased CREs—both H3K27me3 sites and DHS—in CAST but not B6 male liver (Fig 7B). These strain-specific, sex-dependent CREs are found at the Moxd1 promoter and at enhancer regions e1 and e2. Importantly, many but not all the CAST-specific, male-biased CREs at enhancer e2 contain strain-specific SNPs or Indels (Fig 7B; red asterisk, regions lacking such SNPs/Indels). Two downstream enhancer regions, e3 and e4, contain male-biased CREs in both CAST and B6 liver, and may account for the basal level of male-biased Moxd1 expression seen in B6 liver. Positive regression coefficients for the CAST genetic background were seen in both male and female DO mice; however, the effect is stronger in males (+1.68 in males vs. +0.89 in females; Fig 7A). In a corresponding example of a female-biased gene, Bmper, we observed strong activation in CAST female liver (category #8 eQTL) associated with multiple nearby female-biased CREs (S11 Fig). Two female-biased intronic enhancers appear to be sufficient to maintain a 5-fold female bias in the expression of Bmper seen in B6 mouse liver; however, the activation of 9 additional female-biased CREs in CAST liver increased the magnitude of female bias to 17.4-fold in CAST liver (S11 Fig).
S12 Fig presents a case where multiple CREs at or near a cluster of three male-biased genes (Rassf3, lnc9349, lnc9351) with overlapping eQTLs coordinately lose male bias due to the activation of their expression in CAST female liver (category #3 eQTL). This genomic region contains multiple male-biased CREs in B6 liver, all but one of which show increased enhancer activity (H3K27ac marks and/or DHS) in CAST female liver, with no change in CAST male liver. Thus, there is a coordinated loss of nearly all male-biased CREs over a wide (~35 kb) genomic region in CAST liver, driving the increased expression seen in the CAST females (S12B Fig). This effect is strongly sex-dependent and strain-dependent, with strong positive regression coefficients seen in CAST female but not CAST male liver for all 3 genes (S12C–S12E Fig). In this case, however, we also need to consider non-strain-specific SNPs/Indels between CAST and B6 mice in the eQTL region, insofar as genetic effects were also observed in a second strain, PWK, and where the promoter region of lnc9349 includes a sex and strain-dependent CRE that harbors two SNPs that are shared between CAST and PWK mice but absent in B6 mice (S12B Fig, red arrows).
Finally, we present a ~125 kb genomic region within a single TAD on chr 10 with overlapping eQTLs for three female-biased genes, which all lose their sex-biased expression in either B6 liver (Gm4794 (Sult3a2)) or CAST liver (Sult3a1 and Rsph4a), albeit by different mechanisms (Fig 8). Gm4794 (Sult3a2) and Sult3a1 are both regulated by category #2 eQTLs (loss/decrease in female bias due to gene activation in male liver; c.f., higher regression coefficients in DO male liver than DO female liver, whereas Rsph4a is regulated by a category #4 eQTL (loss of female bias by repression in female liver) (S13A–S13C Fig). Gm4794 (Sult3a2) and Sult3a1 both show striking increases in CREs (both H3K27ac marks and DHS) and in gene expression in CAST livers from both sexes, which introduces a sex bias in expression in the case of Gm4794 (Sult3a2) and abolishing one in the case of Sult3a1 (Fig 8A). The loss of female-biased expression of Sult3a1 occurs, even though many of the strain-specific CREs within its eQTL region acquire female-biased H3K27ac marks and/or female-biased DHS in CAST liver (Fig 8B). Strikingly, many of the strain-dependent, sex-specific CREs seen in CAST liver do not overlap strain-specific variants (red asterisks, Figs 8B and S13D).
The eQTL regulating Rsph4a is only significant in female DO mouse liver (-1.01 regression coefficient, CAST-specific; S13C Fig), and consistent with that, Rsph4a is strongly repressed in female CAST liver compared to female B6 liver, abolishing its female-specific expression pattern (Fig 8A). The only B6-specific female-biased CRE in this gene region is at the promoter of Rsph4a and overlaps a CAST-specific SNP (S13E Fig). Sequence variants that disrupt TF motifs are rare, and represent only 1–5% of genetic variants [46]; however, this CAST-specific SNP at the female-biased Rsph4a promoter CRE, seen in B6 but not CAST liver, is one such example: it disrupts an HNF6/Cux2 motif within an HNF6 ChIP-seq binding site identified in CD-1 mouse liver [79] and it likely contributes to the observed loss of female-biased Rsph4a expression.
Discussion
We elucidated both genetic and epigenetic factors that contribute to the sex-biased patterns of hepatic gene expression and cis regulatory activity in two evolutionarily distant mouse strains, B6 and CAST. We characterized both strain-shared and strain-unique sex-biased genes, including many lncRNA genes. Strain-shared sex-biased protein-coding genes were found to be highly liver-specific in their expression and biological functions and were regulated by TSS-proximal enhancers, whereas sex-biased genes unique to each strain were significantly less liver-specific and more often regulated by distal enhancers. We also identified hundreds of cis regulatory elements (CREs, primarily enhancers) with a strain-shared sex-bias, and we elucidated the strain-dependent gain or loss of many other strain-specific, sex-biased CREs. A subset of the strain-specific CREs harbored strain-specific SNPs/Indels, which we linked to eQTLs regulating the strain-dependent, sex-biased expression of individual sex-biased genes. In many cases, these eQTL-gene relationships reflected the gain or loss of a single, apparently causal, strain-specific sex-biased CRE. Remarkably, in many other cases we observed a coordinated gain or loss of sex bias across multiple CREs within an eQTL, including CREs that did not contain strain-specific SNPs or Indels, which raises fundamental questions about underlying mechanisms. Thus, by integrating biological data at the genetic, epigenetic, and transcriptomic levels across divergent inbred mouse strains, we harnessed the power of natural genetic diversity to gain novel insights into the biology and genetics of liver sex differences, including the discovery of hundreds of genomic regions implicated in sex-biased gene regulatory mechanisms.
We identified a core set of 144 protein-coding genes whose sex-biased expression is conserved between B6 and CAST mouse liver. Many are liver-specific genes with strong enrichment for molecular function gene ontologies related to liver physiology. The conservation of sex-bias for these genes across the substantial evolutionary distance between CAST and B6 mice suggests they play a fundamental role in the biology of sex differences in the liver and its regulation. One example is the conservation between strains of the sex-biased expression of Bcl6 and Cux2, two GH-regulated transcriptional repressors (TFs) [71,80] that play a critical role in enhancing the sex-bias of many genes in mouse liver [40,41,79]. We also identified 78 strain-shared sex-biased lncRNAs, which may play regulatory roles in the chromatin-bound fraction either in cis or in trans [18,81,82]. Four of these 78 lncRNAs have been implicated in negative regulation of sex-biased protein-coding genes of the opposite sex bias and inverse hypophysectomy response class (lnc630, lnc2937, lnc7423, lnc9000; Sheet B in S1 Table) [17]. In contrast, three other lncRNAs implicated in negative regulation of other sex-biased genes [17] showed sex-biased expression in B6 but not CAST liver, and could contribute to the strain differences in sex-biased gene expression described here.
Three TFs showed sex-biased expression in B6 but not CAST mouse liver: Tox, Trim24 (both female-biased) and Zfp809 (male-biased). All three TFs are subject to GH-regulated expression [13,71,72] and may contribute to the strain differences in sex-biased gene expression via trans regulation. Tox is a regulatory factor in T-cell exhaustion [83] and is a therapeutic target for cancer immunotherapy [84], but its role in gene expression in hepatocytes is unclear. Zfp809 is a KRAB-zinc finger protein implicated in epigenetic silencing of endogenous retroviral sequences [85]. The male bias in Zfp809 expression seen in B6 but not CAST mouse liver could thus impart strain differences for genes where endogenous retroviral silencing is critical. Incomplete silencing of endogenous retroviral sequences contributes to strain differences in spontaneous and drug-induced liver tumorigenesis [86] but has not been explored in the context of sex differences. Trim24 is a suppressor of mouse hepatocellular carcinoma [87,88]; consequently, its elevated expression in female compared to male B6 mouse liver may contribute to the lower incidence of liver cancer in females [70,89]. Our finding that the female bias in Trim24 expression is lost in CAST mouse liver, where Trim24 shows high expression in males (S2 Fig), suggests sex differences in hepatic tumorigenesis may be reduced in CAST mice. B6 and CAST mice differ in their sensitivity to genotoxic damage induced by 1,3-butadiene, which induces strain-dependent epigenetic and transcriptomic changes in liver, lung and kidney [90,91]; however, differences in hepatocellular carcinoma between mouse strains [92,93] have not been determined for B6 vs CAST mice.
We recently identified a large number of eQTLs that regulate the expression of sex-biased gene expression in mouse liver [49], but little is known about how these autosomal genetic factors impart their regulatory effects. Adding to the problem, the loci identified are large and often contain thousands of SNPs and Indels. Here, we show that many of these eQTLs can be linked to the strain-specific gain or loss of sex-biased chromatin opening and activating enhancer marks at CREs within the eQTL regions, a subset of which harbor strain-specific SNPs/Indels. Thus, 208 of 286 eQTLs with apparent sex-differential effects on sex-biased gene expression showed a complete gain, loss, or reversal of sex-biased expression for the regulated gene. Further, 166 of the eQTLs were associated with a gain or loss of at least one sex-biased CRE harboring strain-specific genetic variants. Importantly, we found that 61 of the eQTL regions contain a single relevant CRE with genetic variants that impact sex-biased expression, which has a high likelihood of being causal for the sex-dependent effects of the eQTL on sex-biased gene expression. Further, for 105 other eQTLs, strain differences in expression were linked to a coordinated gain or loss of multiple strain-dependent, sex-biased CREs within the eQTL region. While we cannot conclusively distinguish a primary, causal enhancer CRE from secondary CREs within a given eQTL based on our study design, in many cases the relevant strain-specific CREs lacked strain-specific genetic variants, but nevertheless showed a gain or loss of sex-bias coordinated with other, neighboring CREs harboring strain-specific variants, indicating they are likely responding in a secondary manner to a causal, strain-specific genetic event.
It is a major challenge to ascertain the contributions of the individual, constituent CREs, and any causal association with gene expression, for eQTL regions that contain multiple CREs with strain-specific SNPs. For such eQTLs, genetic variants within one of the multiple eQTL CREs may be the primary causal driver of the observed gain or loss of sex-biased enhancer activity and gene expression across the eQTL. For example, a SNP/Indel that activates a single causal, strain-specific CRE may nucleate activity at neighboring CREs that lack an activating sequence variant, as occurs in T-cell leukemias, where a mutation at a single oncogenic enhancer activates multiple other enhancers and up regulates the TAL1 oncogene [94]. Cooperativity in enhancer activation of proximal genes is well established [95], however, the exact nature of the relationship between clusters of enhancers and gene targets remains controversial [96]. In some cases, deletion of single enhancers leads to a dramatic loss of activity at neighboring enhancers, suggesting a hierarchical relationship [94,97,98], with a clear delineation between hub and non-hub [99] and between predominant and supportive enhancers [98] within larger enhancer clusters. However, other studies find that enhancers contribute mostly additively to gene expression, with redundancy between individual enhancers [100,101]. Indeed, there may be fundamental biological differences between enhancer clusters, only some of which exhibit hierarchical 3-dimensional structure [99]. Even within a single enhancer cluster, hierarchical relationships may vary, depending on hormonal cues and downstream signaling [97,102]. Further work, including careful dissection using genetic editing in mouse liver in vivo, will be needed to determine the respective contributions of individual sex-biased CREs whose strain-dependent gain or loss of activity is coordinated with neighboring regulatory elements, and to elucidate the underlying features that determine whether a given eQTL region best fits a hierarchical model or an additive model of enhancer coordination.
None of the strain-specific genetic variants presented here, except one at the eQTL for Rsph4a, directly disrupts a TF motif. Indeed, we found that strain-specific genetic variants are depleted from intra-motif coordinates relative to randomized genomic coordinates, consistent with prior reports [45,46,77,78]. Disruption of enhancer activity may occur by a combination of effects on DNA shape, nucleosome positioning, and co-binding TFs [103]. Liver sexual dimorphism requires the coordinated participation of multiple TFs, both those that are sex-biased and/or GH-responsive, such as STAT5 [40,41,57,79], and sex-independent TFs essential for many liver-expressed genes and functions [37,70]. This coordination is likely to involve TF-TF interactions within large, co-bound TF clusters, which are stabilized by multiple suboptimal motif-adjacent sequences [104]. While genetic variants outside of canonical binding motifs may have modest effects on the binding of individual TFs, they may nevertheless destabilize the overall multi-TF clusters that form [105]. Consistent with this idea is the coordinated gain or loss of multiple strain-specific, sex-biased CREs that we found at 105 eQTLs regulating the strain-specific expression of sex-biased genes.
Our comparison of sex-biased gene expression and sex-biased regulatory elements across mouse strains has some limitations. We investigated the impact of strain-specific genetic variation on the activity of CREs, but did not consider genetic differences at transcribed sequences, which can impact RNA stability [106]. Furthermore, genetic variants may contribute to differences in 3D genome organization [107–109], which may differ between B6 and CAST mouse liver. Our mapping of sex-biased CREs to eQTL target genes has already taken into account genome structure in the form of TADs, which define broad regulatory domains that restrict enhancer-promoter interactions [110,111] and significantly enriched for genetic variants at sex-biased CREs as compared to full-length eQTL regions (Fig 4B). However, we did not consider potential strain differences in TAD or intra-TAD structure [76,112,113], which may allow cis regulation to be defined with greater precision [74]. Moreover, strain differences in DNA looping may be independent from the strain differences in epigenetic marks of enhancer activity that we measured, and consequently, some of the strain-shared sex-biased CREs identified here may interact with their sex-biased gene targets in a strain-specific manner. Chromosome conformation assays have revealed sex-dependent looping interactions in mouse liver [54] and may be useful for evaluating strain-specific sex bias at the level of DNA looping.
The nomenclature of “gain” and “loss” of sex bias used throughout this study is based on the underlying assumption that the private/unique genetic variants that are the focus of this study are unlikely to be shared between the regulating strain and the ancestral state (see Methods for additional definition of these terms). While we consider this assumption most probable, it remains a possibility that the ancestral allele and even an allele that is strain-specific among DO founder strains are shared. This is more likely for CAST mice than for B6 mice, given its evolutionary distance from the other founder strains. Further analysis of several additional wild-derived mouse strains more closely related to CAST would be required to further evaluate this issue.
Our work is also limited by its focus on only two mouse strains, B6 and CAST. In particular, by focusing this study on strain-specific genetic variants and by strictly requiring that these variants be unique across all 8 DO founder strains, our analysis explicitly excludes variants shared between B6, or CAST, and one or more other founder strains. Of note, this exclusion has the beneficial effect of greatly limiting the number of SNPs to be considered within an eQTL region (e.g., Fig 5, Unique vs All variants track). Moreover, this approach is appropriate for the 70–74% of eQTLs that do not involve major contributions from a second strain (125 of the 168 robust sex-biased eQTLs; and 344 of all 491 eQTLs; Sheet A in S6 Table), and as a result, we consider it less likely that non-strain specific SNPs account for the strain differences in expression between B6 and CAST mice for these eQTLs. Further, as noted above, we cannot validate our assignments of loss and gain of sex-specific gene expression alleles without knowing the true ancestral allele. A loss of gene expression pattern in one strain could be a gain of gene expression pattern in multiple DO founder strains, a case more likely to occur when the mutation is acquired prior to a speciation event, for example, speciation of the six M. m. domesticus strains examined here.
Finally, while a majority of intra-population variation in TF binding or chromatin state is attributable to cis-acting genetic variants [46,114], genetic factors may also impact sex-dependent gene expression through strain-specific trans-acting factors [115]. Indeed, we identified 41 trans-acting eQTLs that affect sex-biased gene expression, nine of which target sex-biased lncRNAs (Sheet A in S6 Table), which themselves may act in a trans manner. In addition, a subset of the strain-unique, sex-specific CREs that we identified is likely not the direct result of local genetic differences, but rather may reflect a loss in CAST mouse liver of sex differences in the expression of their bound transcriptional regulators, such as Tox, Zfp809 or Trim24, as noted above. An F1 cross of CAST x B6 mice could address this question by placing a copy of each genome in the same trans environment, and thereby help differentiate cis from trans effects on gene expression and transcription factor binding [46,51]. Furthermore, it is difficult to ascertain lineage-specific gain or loss of sex-bias without an outgroup such as M. Spretus (~2MY diverged from CAST and B6 mice; [116]), for which gene expression and CRE data (TF binding activity) is only available for male liver [45]. Thus, the terms gain and loss of sex-bias, as used here, may not represent the full evolutionary history of murine subspecies.
In conclusion, we report strong genetic effects on the expression of sex-biased genes in a mouse liver model, with several hundred eQTLs resulting in a complete gain, loss, or reversal of sex-biased expression between B6 and CAST mice. We identified at least one CRE that gains or loses sex bias (either chromatin opening or H3K27ac mark accumulation) in a manner that can explain the gain or loss of sex-biased gene expression for a majority of these eQTLs. In many cases, we observed the coordinated gain or loss of sex bias across multiple CREs within an eQTL, which makes the elucidation of causality of individual genetic variants difficult. Despite the widespread occurrence of these strong genetic factors, we identified a core set of strain-shared sex-biased genes, including many lncRNA genes, with liver-specific patterns of expression and molecular functions. We can anticipate that our findings, and the approaches taken in this investigation of the role of genetics and epigenomics in liver sex differences, will be critical for translating studies from mouse models to humans.
Methods
Ethics statement
Mice were treated using protocols specifically reviewed for ethics and approved by the Boston University Institutional Animal Care and Use Committee (protocol approval # PROTO201800698), and in compliance with ARRIVE 2.0 Essential 10 guidelines [117], including study design, sample size, randomization, experimental animals and procedures, and statistical methods.
Animals
Adult male and female C57BL/6J (B6) and CAST/EiJ (CAST) mice were purchased from Jackson Laboratories (cat. # 000664 and # 000928, respectively; Jackson Laboratories, Bar Harbor, ME) and were housed in the Boston University Laboratory Animal Care Facility (n = 5 per sex for each strain; lights on 7:30 am to 7:30 pm). Mice were euthanized at 8 weeks of age by cervical dislocation, and livers were removed then rinsed in ice cold PBS. For consistency and to minimize the impact of circadian rhythms [118], all mice were euthanized between 10:45 and 11:30 am. Batch effects were minimized for both tissue collection and downstream experiments by including in each group of mice or tissue samples processed, biological replicates from each strain and sex (i.e. male CAST, female CAST, male B6, and female B6 mice). Thus, when collecting livers, no more than two biological replicates for each combination of strain and sex were processed on a given day. A portion of each liver was snap frozen and processed later for RNA isolation (~10% of each liver), or by crosslinking and ChIP-seq (40% of the total mass). The remaining ~50% of each liver was directly used for nuclei isolation (see below).
RNA isolation and library preparation
RNA was extracted from frozen liver using TRIzol reagent (Invitrogen, cat. # 15596026) following the manufacturer’s directions. Total RNA was quantified using the Qubit RNA BR kit (Invitrogen, cat. # Q10210). One μg of total RNA per sample was used as input for library preparation using the NEBNext Ultra Directional RNA Library Prep kit for Illumina (NEB, cat. # E7420) with the PolyA selection with the NEBNext Poly(A) mRNA Magnetic Isolation Module (NEB, cat. # E7490). Agencourt AMPure XP beads (Beckman Coulter, cat. # A63881) were used to remove low molecular weight material prior to final quantification of purified libraries using the Qubit dsDNA HS kit (Invitrogen, cat. # Q32854).
Isolation of nuclei, DNase I digestion and library preparation
Nuclei, isolated from freshly excised liver were lightly digested with DNase I to nick accessible chromatin [119], with modifications [120]. Batch effects were minimized by processing 6 samples in parallel but including no more than 2 biological replicates of the same strain and sex in each group of nuclei. Briefly, livers were homogenized in a Potter-Elvehjem homogenizer using high sucrose homogenization buffer (10 mM HEPES (pH 7.5), 25 mM KCl, 1 mM EDTA, 2 M sucrose, 10% glycerol, 0.05 mM DTT, 1 mM PMSF, 0.15 mM spermine, 0.2% (v/v) spermidine, 1 mM Na orthovanadate, 10 mM NaF, and 1X Roche Complete Protease Inhibitor Cocktail, cat. # 11697498001) to prevent aggregation of nuclei and preserve chromatin structure. The resulting slurry was placed on top of a 3 ml cushion of homogenization buffer followed by ultracentrifugation at 25,000 RPM for 30 min at 4°C in an SW41 Ti rotor to pellet the nuclei and remove cellular debris. The supernatant was carefully decanted to remove liquid, and residual solid debris was removed from the tube walls using a sterile spatula and a dampened Kimwipe. Nuclei were resuspended in 1 ml of Buffer A (15 mM Tris-HCl pH 8.0, 15 mM NaCl, 60 mM KCl, 1 mM EDTA pH 8.0, 0.5 mM EGTA pH 8.0, 0.5 mM spermidine, 0.3 mM spermine) and transferred to a 1.5 ml microcentrifuge tube. Nuclei were taken directly for DNase treatment at this point.
Nuclei were counted using an Invitrogen Countess instrument (cat. # C10281), and 50 million nuclei per liver were incubated in a volume of 950 ul with 32 U of RQ1 RNase-free DNase I (Promega, cat. # M6101) for 2 min at 37°C. To extract the released DNA fragments, samples were extracted with phenol/chloroform then layered on a sucrose gradient, followed by centrifugation at 25,000 rpm for 24 h at 25°C. Gradient fractions (1.9 ml each) were collected, and fractions enriched in DNA fragments between 0.1 and 1 kb in size (fractions 7–11; numbered from the top-most fraction) were pooled and then purified by double-sided SPRI bead selection using Agencourt AMPure XP beads to obtain DNA fragments between 125–400 bp long. DNA was quantified using the Qubit dsDNA HS kit and further qualified prior to library preparation by qPCR using primers previously shown to amplify open chromatin regions in mouse liver [36,119]. DNase libraries were prepared for sequencing using NEBNext Ultra II DNA Library Prep Kit for Illumina following the manufacturer’s instructions (NEB, cat. # E7645), with double-sided SPRI size selection (Agencourt AMPure XP beads) to obtain an average insert size of 200 bp prior to PCR amplification, and single-sided SPRI after PCR amplification to remove low molecular weight material.
Chromatin crosslinking, immunoprecipitation, and library preparation
Chromatin was extracted from the ~40% snap-frozen, cross-linked liver aliquots as described [121], with modifications. Batch effects were limited by including no more than 2 biological replicates from the same strain and sex in each set of six liver samples processed together in a single batch. Briefly, livers were resuspended in a total volume of 4.687 ml of XL Buffer (50 mM Hepes, pH 7.5, 100 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 1X Roche Complete Protease Inhibitor Cocktail), processed with a Dounce homogenizer, and passed through a 70-micron cell strainer (Corning, cat. # 352350). 313 μl of 16% formaldehyde (Pierce, cat. # 28906) was then added and the samples were nutated for 10 min at 22°C. Crosslinking was halted by the addition of 250 μl of 2.5 M glycine, followed by additional nutation for 2 min at 22°C, then incubation on ice for a minimum of 5 min. Cells were pelleted at 2,500 g for 5 min at 4°C, the supernatant was discarded, and the pellets were rinsed twice with ice cold PBS containing 1X Roche Complete Protease Inhibitor Cocktail, pelleting between each wash as above. The cell pellet was then lysed by resuspension in 10 ml of Lysis Buffer-1 (50 mM Hepes, pH 7.5, 140 mM NaCl, 1 mM EDTA, 10% glycerol, 0.5% NP40, 0.25% Triton X-100) followed by nutation for 10 min at 4°C. Lysed cells were pelleted, as above, and nuclei were lysed by resuspension in 10 ml Lysis Buffer-2 (10 mM Tris-HCl pH 8.0, 200 mM NaCl, 1 mM EDTA, 0.5 mM EGTA) followed by nutation for 5 min at 4°C. Crude chromatin extract was pelleted, as above, and resuspended in 2 ml of 0.5% SDS RIPA buffer (50 mM Tris-HCl, pH 8.0, 150 mM NaCl, 1% IPEGAL, 0.5% deoxycholic acid).
Chromatin was transferred to a 15 ml conical tube (Diagenode, cat. # C30010017) for sonication in a Bioruptor Pico instrument (Diagenode, cat. # B01060010). Sonication was performed at 4°C for 25 cycles (30 s ON and 30 s OFF at high intensity) with 2 min rests on ice every 10 cycles to prevent sample heating. A small aliquot of this material was reverse crosslinked (65°C for 6 h), treated with RNase A (5 μg per sample at 37°C for 30 min; Novagen, cat. # 70856) and then digested with proteinase K (20 μg per sample at 56°C for 2 h; Bioline, cat. # 37084) and electrophoresed on a 1.5% agarose gel to verify that the majority of fragments were 100–400 bp in size. This sonicated material was quantified using PicoGreen Quant-iT dsDNA BR (Invitrogen, cat. # P11496). Prior to antibody incubation (below), ~5% of chromatin by volume was removed and used as a background, pre-immune, input control (total of n = 4 control samples, one per strain per sex).
Immunoprecipitation of mouse liver chromatin was performed as described [52]. Protein A Dynabeads (15 μl; Invitrogen, cat. # 1002D) were incubated in blocking solution (0.5% bovine serum albumin in PBS) with 3 μg of antibody to histone-H3 K27ac (Abcam, cat. # ab4729) for 3 h at 4°C. Bead immune-complexes were washed with blocking solution, followed by overnight incubation with 15 μg of liver chromatin diluted to a final concentration of 15 ng/μl in RIPA buffer containing 0.1% SDS. After washing with 1X RIPA buffer (containing 0.1% SDS) and reverse crosslinking as described above, DNA was purified using the QIAquick Gel Extraction Kit (Qiagen #28706) and quantified using the Qubit dsDNA HS kit. ChIP-seq libraries were prepared for sequencing using NEBNext Ultra II DNA Library Prep Kit for Illumina, with double-sided SPRI size selection to obtain an average insert size of 200 bp prior to PCR amplification, and single-sided SPRI after PCR amplification to remove low molecular weight material.
qPCR analysis, pooling of libraries and Illumina sequencing
The quality of all ChIP and DNase-digested samples was validated prior to preparation of sequencing libraries, as follows. qPCR analysis was carried out using primers (Sheet A in S4 Table) for three sex-biased genomic regions, for one sex-independent positive control DHS peak region, for one sex-independent positive control peak region (an H3K27ac-positive site), and for three negative control regions. DNase-qPCR data for sex-biased DHS regions are presented (S5A and S5B Fig) as signal to noise, based on the average of three negative control (background) regions, calculated as follows: 2^[(DHSgenomic−DHSDNase)/(NegAvg)], where NegAvg = [(NEG1genomic−NEG1DNase)+(NEG2genomic−NEG2DNase)+(NEG3genomic−NEG1DNase)]/3. “DHS” indicates data obtained with a qPCR primer pair within the DHS region and “NEG” indicates 1 of 3 primer pairs specific to the negative control regions. A subscript “DNase” indicates CT (qPCR cycle threshold) values for qPCR reactions using DNaseI-digested DNA as template, while a subscript “genomic” indicates CT values for qPCR reactions using genomic DNA as template. ChIP-qPCR data (S5C Fig) is presented as percent input, calculated as follows: [100/2^(CT value for ChIP DNA–CT value for input DNA)] x [(final volume of ChIP sample) x (initial volume of input sample)] / [(initial volume of ChIP sample) x (final volume of input sample)]. ChIP and DNase-digested sample quality was determined using sex-independent and negative control primers. Samples with the highest signal to noise (Positive Region % input) / (Avg of 3 Neg Ctl region % Input) for ChIP samples; and as described above for DNase-digested samples) were sequenced, as described below.
All sequencing libraries were multiplexed with a dual-indexing strategy using NEBNext Multiplex Oligos for Illumina (NEB, cat. # E7600). Final libraries (n = 5 biological replicates per sex and per strain; 20 libraries each for total liver RNA-seq (polyA-selected), DNase-seq, and H3K27ac ChIP-seq, plus a total of 4 ChIP-seq input control samples (64 libraries in total) were quantified in a single batch using the Qubit dsDNA HS kit. ChIP-seq, RNA-seq, and DNase-seq libraries were pooled on an equimolar basis and sequenced (PE-50 reads) on an Illumina NovaSeq 6000 instrument at the New York Genome Center to an average depth of 34 million read pairs per sample (mean Q ≥ 30 for 98.0% of Read-1 and 95.7% of Read-2). We excluded from downstream analysis one of the n = 5 biological replicate DNase-seq libraries per group, due to variable sample quality, as confirmed by qPCR analysis of a sex-independent positive control DHS peak region.
Custom CAST mouse genome
The B6 mouse genome (mm9) was converted to a custom genome incorporating known genetic variants with g2gtools (v0.2.7; https://github.com/churchill-lab/g2gtools), using quality SNPs/indels downloaded from the Sanger Mouse Genome project, Release 1211 [61]. First, the vcf file from Sanger was converted to an indexed vci file, using the command: "g2gtools vcf2vci—quality -o CAST.vci -s CASTEiJ -p 8 -i mgp.v2.indels.annot.reformat.withchr.vcf.gz -i mgp.v2.snps.annot.reformat.withchr.vcf.gz -f NCBIM37_um_all.fa”. Second, SNPs were patched into the mm9 genome using the command: “g2gtools patch -i NCBIM37_um_all.fa -c CAST.vci.gz -o CAST_patched.fa -p 8”. Third, Indels were incorporated using the command: “g2gtools transform -i CAST_patched.fa -c CAST.vci.gz -o CAST.fa -p 8”. Finally, to annotate the custom CAST genome, custom CAST mouse GTF files were generated for protein-coding gene and lncRNA gene locations converted from mm9 to the newly-generated custom CAST genome using the command “g2gtools convert -i merged_refseq_ncRNA.gtf -c CAST.vci.gz -o merged_refseq_ncRNA.gtf” (for RNA-seq). General conversion of mapped reads, peak lists, or other files with CAST genomic coordinates to B6 (mm9) coordinates was performed using the command: “g2gtools convert -i test_CASTcoord.bed -c CAST.vci.gz -o test_mm9coord.bed -f bed–reverse”. CAST-derived sequence reads mapped to the custom CAST mouse genome showed mapping frequencies and cumulative gene expression very similar to B6 mice (Sheet B in S4 Table).
Mouse liver RNA-seq samples and differential expression analysis
Microarray datasets included in S4E Fig are based on data generated by Jackson Labs and analyzed previously (http://cgd.jax.org/gem/strainsurvey26/v1) [49]. RNA-seq data for CD-1 mice was download from GSE93380 (samples GSM2452246-GSM2452251), and represents three RNA pools per sex, each comprised of polyA-RNA from n = 12 to 17 individual livers per sex [13]. For B6 and CD-1 mouse liver RNA-seq datasets, reads were mapped to the mm9 genome using TopHat (v2.1.1) [122] with default parameters. For CAST mice, sequence reads were mapped to the custom CAST genome (described above) using TopHat. The gene annotation file used for both strains was identical (prior to coordinate liftover) and included both coding and noncoding genes, as described in [16,123] and implemented in [49]. Mapped reads were filtered to include only primary, uniquely mapping reads, which were assigned to gene-level features based on overlap of at least one nucleotide with any exon using FeatureCounts (v1.6.2) [124]. Reads counts were transformed to fragments per kilobase of exon per million reads mapped (FPKM) for all downstream analysis and visual comparisons. Differentially expressed genes were identified using EdgeR (v3.8) [125] with default parameters. Genes differentially expressed between the sexes were identified based on a male liver vs female liver |fold change| > 2 (i.e., sex bias >2 fold) at an FDR < 0.05 (i.e., EdgeR (Exact test) adjusted p-value < 0.05). We defined strain-unique (strain-specific) sex-biased genes as genes that met the threshold of sex bias > 2 and FDR < 0.05 in either B6 or CAST mouse liver but failed to meet the FDR cutoff in the other strain (FDR > 0.05). Strict strain-shared sex-biased genes met the above sex bias and FDR cutoffs in both strains, while standard strain-shared sex-biased genes met the FDR cutoff in both strains, but the sex bias fold-change cutoff in only one strain. These definitions were without regard to a gene’s sex bias in CD-1 mouse liver.
Mouse liver ChIP-seq and DNase-seq samples and epigenetic data analysis
For B6 mice, ChIP-seq and DNase-seq reads were demultiplexed and mapped to the mouse genome (build mm9) using Bowtie2 (v2.2.9) [126], allowing only uniquely mapped reads. For CAST mice, sequence reads were mapped to the custom CAST genome, allowing only uniquely mapped reads. Peaks of sequencing reads were identified using MACS2 (v2.1.1) [127] as regions of high signal over the background input DNA samples. Peaks were filtered to remove blacklisted genomic regions (www.sites.google.com/site/anshulkundaje/projects/blacklists). Genomic regions called as peaks but that contain only PCR duplicated reads, defined as > 5 identical sequence reads that do not overlap any other reads, were also removed. Differential peaks were identified by diffReps (v1.5.6) [73] with default parameters, then filtered to require a fold-change > 2 (or fold-change > 1.5 for DNase-seq). To account for sample-to-sample variability and increase robustness, we retained for downstream analysis those differential regions that overlap a peak region identified by MACS2, and with a minimum read count of n = 15 for ChIP-seq samples and n = 10 for DNase-seq samples.
Genomic coordinates for reads, peaks, and signal (BigWig) mapped to the custom CAST genome were converted to mouse mm9 coordinates for visualization and normalization. All BigWig tracks used for genome browser visualization were normalized for sequencing depth and fraction of reads in peaks, expressed as reads in peaks per million mapped reads (RIPM) using Deeptools2 (v2.3.3) [128]. Read in peak normalization and diffReps filtering were performed relative to the union of peaks across all samples (B6 and CAST). All browser screenshots are generated using the WashU Epigenome Browser (https://epigenomegateway.wustl.edu). Screenshots presented in the figures are based on samples merged by strain and sex at the fastq level and processed as described above (i.e., all n = 5 male B6 H3K27ac ChIP-seq fastq files were merged; and the same was done for B6 female, CAST male, and CAST female samples). Each dataset was visualized with a pair of tracks representing the signal (top; BigWig file normalized as above) and peak/DHS positions (bottom; bed file described below). The signal track superimposes merged male (blue) and merged female (pink) replicates (merge of n = 5 for H3K27ac ChIP-seq samples and merge of n = 4 DNase-seq samples; above) in a single track (matplot function of WashU browser). H3K27ac peak and DHS positions are shown for sex-biased peaks and DHS regions (Sheets E-H in S5 Table) and for sex-independent robust peaks and DHS regions (Sheets B and D in S5 Table).
Proximity of genes to CREs
In Fig 3A-C, distance from the TSS of genes on an indicated gene list (S1 Table) to the nearest CRE on the indicated peak list (S5 Table) was calculated using bedtools (closest -d; bedtools v.2.28.0). For the stacked bar chart in Fig 3D, we assumed that core (strain-shared) sex-biased genes could be regulated by neighboring CREs in one of two ways: 1) sex-biased DHS or K27ac mark accumulation that is sex biased in both strains at the same site (i.e. 533 common CREs; Sheets E and F in S5 Table) or 2) sex-biased DHS or K27ac mark accumulation that is sex biased at a different site in each strain, both of which meet the criteria for the classification (i.e. both a CAST-specific and B6-specific CRE at different sites regulating the same gene; Sheets G and H in S5 Table). For example, a gene was considered proximally-regulated if the TSS of that gene had a CRE within 20 kb that met the criteria described in scenario 1 or 2, above.
Assigning eQTL and genetic variants to genes
Previous work from this lab identified 1,172 eQTLs associated with the expression of one or more of 1,033 sex-biased genes [49]. For 414 of these eQTLs, the regulating strain in at least one condition is either CAST or B6 (i.e., based on analyses considering either all 438 DO mouse liver samples, 219 male DO liver samples only, or 219 female DO liver samples only). As discussed in [49], for CAST liver, these eQTLs only considered sex-biased genes identified by microarray, which is less sensitive and less specific than RNA-seq and largely limited to protein coding genes (“TRUE” in column AF of Sheet A in S6 Table). To address this issue, we additionally considered any eQTL associated with CAST-specific sex-biased genes (Sheets D and F in S1 Table), which yielded an additional 77 eQTLs (“FALSE” in column AF of Sheet A in S6 Table), giving a final list of 491 eQTLs for which CAST or B6 is the regulating strain associated with 481 sex-biased genes. A separate listing of genes showing sex-biased expression in CAST liver is shown in Sheet C in S6 Table, including annotation for genes not previously identified as sex-biased.
Definitions for regulating strain and gain or loss of sex bias
The term “regulating strain” is used to indicate the strain in which an eQTL was identified that has the most significant LOD score. The terms “gain” and “loss” of sex bias in the regulating strain are based on the assumption that any private genetic variant (i.e., one unique among all DO founder strains) differs from the ancestral allele.
Contributing genetic variants
The following strategies were employed to narrow the genomic region under consideration for putative contributing genetic variants. First, for eQTLs significant in >1 set of DO liver samples with B6 or CAST as the regulating strain, we considered up to 3 datasets giving significant eQTLs: all 438 DO liver samples, 219 DO male liver samples only, and 219 DO female liver samples only. We selected the shortest eQTL region of the three (defined as the 95% Bayesian credible interval; [49]). If the eQTL was only significant in B6 or CAST mice in one of these liver sets, then those coordinates were chosen (by default it is the shortest). These values are shown in columns A and B of Sheet A in S6 Table. For eQTLs significant in more than 1 of the 3 sets mentioned above, we determined the intersection of the 2 (or the 3) significant genomic intervals as a secondary set of genomic coordinates, which are listed in columns D and F of Sheet A in S6 Table. Finally, for eQTL intervals that overlap with the TAD containing the TSS of the regulated gene (column Y = “tad”), the intersection of the eQTL and TAD coordinates is shown in columns G-I of Sheet A in S6 Table. TAD boundaries were those defined for male B6 mouse liver [74,129].
Discovery of robust sex-biased eQTLs by explicitly modeling sex-QTL interaction
We used the approach of [75] to investigate the hypothesis that eQTLs may have a different effect in each sex. Briefly, the effect of sex on eQTLs was assessed by building two models: Model_A, which includes sex, DO generation, genotyping batch, diet, and sex*diet as additive covariates; and Model_SexInteraction, which in addition to the additive covariates in Model_A, also includes sex as a covariate that interacts with the eQTL (eQTL x sex interaction). The difference of LOD score between the two models (ΔLOD) was used as an estimate of the eQTL x sex interaction, i.e., how different the impact of the eQTL is in each sex. We determined the significance threshold for ΔLOD by performing 1000 permutations of shuffling genotypes and phenotypes for both Model_A and Model_SexInteraction. To ensure that both permutations used the matched permutations of data, we set the seed for the random number generator to be identical before each permutation. The distribution of ΔLOD values was used to determine the genome-wide threshold with α of 0.05 (ΔLOD = 4.27). All the above analyses were performed in R/eQTL2 [130]. The resultant set of 168 robust sex-biased eQTLs regulating sex-biased genes is a subset of the full set of 491 eQTLs regulating sex-biased genes described previously [49], and is identified in Sheet A in S6 Table, columns FS-FU.
Categorization of eQTLs based on their impact on sex-biased gene expression
The genetic effects uncovered by eQTL analysis in DO mice in relation to the regulating founder strain was described previously [49]. For example, in cases where eQTL analysis in male DO mice yielded an eQTL with a negative regression coefficient, and with CAST as the regulating strain, the gene target of this eQTL is referred to as repressed in CAST male liver. Of the full set of 491 eQTLs regulating sex-biased genes in liver with CAST or B6 as the regulating strain, 355 eQTLs were associated with genes that are significantly sex-biased in B6 and/or CAST mouse liver and 388 eQTLs can be categorized into one of 8 categories described below (Sheets A, column X, and G in S6 Table). The sets of robust sex-specific and strain-specific eQTLs are annotated in Sheet A in S6 Table (columns FU and FV) and in Sheet G in S6 Table (column N). The number of eQTLs that can be categorized (388 of 491) is less than 491 because we only considered eQTLs that are significant in the DO mouse male-only liver set (n = 219), or in the DO mouse female-only liver set (n = 219), and for which CAST or B6 is the regulating strain, or contributes substantially to the regulation as indicated by a regression coefficient within 20% of the absolute maximum regression coefficient in the regulating strain, as determined in [49]. The intersection of the 355 eQTLs regulating genes showing sex-biased expression in either B6 and/or CAST livers with the 388 categorized eQTLs resulted in 286 eQTLs (S9G Fig). eQTLs were categorized based on three factors: the sex-bias of the regulated gene, whether the eQTL was stronger in male or in female liver (by LOD score), and the directionality of the effect on regulated gene expression (positive or negative regression coefficients) (Fig 4C). The sex-bias of the gene was assigned based on the absolute maximum Male/Female log2 fold-change across DO founder strains showing significant sex-biased gene expression (column AJ of Sheet A in S6 Table). Negative values in column AJ of Sheet A in S6 Table indicate female-biased genes (eQTL categories #2, 4, 6 or 8) and positive values indicate male-biased genes (eQTL categories #1, 3, 5 or 7). Next, if the LOD score in DO male livers was higher than in DO female livers (positive value in column AS of Sheet A in S6 Table) then the eQTL was considered “stronger in male” (categories #1, 2, 5 or 6), and if the LOD score in DO females was higher than in DO males then the eQTL was considered “stronger in female” (categories #3, 4, 7 or 8). Finally, we considered the impact on regulated gene expression as reflected by the regression coefficients in the sex with the stronger eQTL (columns CD-CT and DD-DT of Sheet A in S6 Table). In the sex with the stronger eQTL, a high positive coefficient represents activation of the regulated gene (categories #2, 3, 5 or 8) while a low negative coefficient represents repression of the regulated gene (eQTL categories #1, 4, 6 or 7; Fig 4C).
Impact on gene expression (Fig 4D) was calculated for the 286 eQTLs regulating genes with significant sex-bias in either B6 or CAST mouse liver. For eQTL categories #1–4, a loss of sex-bias was defined as significant sex-bias in the non-regulating strain in combination with no significant sex-bias in the regulating strain (where significant was defined as EdgeR FDR < 0.05; both here and elsewhere in this section). For eQTL categories #5–8, a gain of sex-bias was defined as a significant sex-bias in the regulating strain and no significant sex-bias in the non-regulating strain. For eQTL categories #1–8, a reversal of sex-bias was defined as a significant sex bias in both regulating and non-regulating strains but with opposite directionality. For categories #1–8, sex-bias retained was defined as significant sex-bias in both the regulating and the non-regulating strains, and with the same directionality, but at either a reduced magnitude (eQTL categories #1–4) or an enhanced magnitude (eQTL categories #5–8). The directionality of sex-bias is shown in columns Z (B6) and AA (CAST) of Sheet A in S6 Table, and significance is shown in columns AG (B6) and AH (CAST). As noted above, the terms loss and gain of sex-specific gene expression pattern used here are predictions based on the observed change of sex-specific gene expression pattern in the regulating strain, as compared to other strains examined, and the specific category of eQTL we identified (categories #1–8). However, absent knowledge of the true ancestral allele, we have no way to verify these assignments. Accordingly, a subset of the alleles that we have designated as a gain of sex-specific gene expression could in fact be a loss of sex-specific gene expression pattern in multiple DO founder strains, and vice versa.
Each eQTL was associated with the DO mouse founder strain showing the highest absolute maximum contributing coefficient. An eQTL was said to be associated with more than one DO mouse founder strains if the contributing coefficient to the eQTL for a second strain was within 20% of that of the strain with the highest absolute maximum coefficient. The number of strains contributing to each eQTL is shown in Sheet A in S6 Table, columns BD, CD and DD; and the results are summarized in column FV.
Gain or loss of sex-biased CREs relevant to eQTL regulation (Fig 4E) was calculated for the 388 eQTLs that can be categorized (Sheet A in S6 Table, column X). Relevant CREs were first defined as those that contain strain-specific genetic variants from the regulating strain and that have the same directionality as the regulated gene (i.e., male-biased gene and male-biased enhancer). For eQTL categories #1–4, relevant sex-biased peaks and DHS were defined as those that lose sex-bias in the regulated strain (i.e., for a gene whose male-biased expression is lost in CAST liver, a B6-specific male-biased peak is considered relevant). For eQTL categories #5–8, relevant sex-biased peaks and DHS were defined as those that gain sex bias in the regulated strain (i.e., for a male-biased gene that gains male-biased expression in CAST liver, a CAST-specific male-biased peak is considered relevant). If the eQTL region included only a single relevant CRE (where, for the purposes of counting CREs, overlapping DHS and K27ac peaks are counted only once), then it was designated ’One’ in Fig 4E. If multiple relevant CREs fell within the eQTL region, or if no relevant CREs fell within the eQTL, then it was designated ’Multiple’ or ’None’, respectively (Sheet A in S6 Table, columns DU and DV).
Strain-specific genetic variants within eQTLs
Both SNPs and Indels were incorporated into the custom CAST genome and were considered as genetic variants. Strain-specific conserved genetic variants were defined as variants that occur in B6 or CAST mice that do not have a genetic variant at the same genomic locus (defined as a single nt in mm9 reference coordinates) in any of the other seven DO founder mouse strains, as described [49]. Strain-specific, non-conserved genetic variants were defined as variants that occur in B6 or CAST mice that do have a genetic variant at the same genomic locus in at least one other DO founder mouse strain, however, the variant is different between the strains (e.g., A -> T in CAST mice and A -> C in PWK mice). Furthermore, only strain-specific conserved and strain-specific non-conserved strain-specific variants were evaluated in this study; we did not consider other genetic variants that distinguish CAST and B6 mice. Genetic variant overlap with CREs was defined using bedtools intersect. Browser screenshots presented in the figures mark only the strain-specific genetic variants within CREs (see S9D Fig). In total, we identified 10,195,846 genetic variants that met our definition of strain-specific in either CAST or B6 mice, of which 124,206 fell within CREs located within eQTL regions (S9D Fig). A B6-specific or CAST-specific variant occurs every 275 bp, on average, calculated based on the size of the mouse genome (2.8 x 109 bp) and the total number of strain-specific variants (10,195,846).
The enrichment of strain-specific genetic variants in features (Fig 4B) was calculated for three sets of features: sex-biased CREs, defined as any sex-biased H3K27ac peak or sex-biased DHS in either strain (Sheets E-I in S5 Table); all CREs, defined as any lenient H3K27ac peak or DHS (Sheets A and C in S5 Table), which also includes robust H3K27ac peaks; and intra-motif, defined as any transcription factor (TF) motif, regardless of the expression level or activity of the TF in liver [131]. The database used represents B6 mouse (mm9) genomic coordinates for all predicted mouse TFs identified in [131] by integrating multiple sequence features into a probabilistic Bayesian model. Overlap was defined as a minimum of 1 bp overlap between a genetic variant in the regulating strain with a TF motif. The number of genetic variants within the indicated feature was calculated for all genetic variants genome-wide, all genetic variants within the 491 eQTLs associated with sex-biased genes, and all genetic variants within the intersection of the 383 intra-TAD eQTLs and the TAD containing the TSS of the regulated gene. This number is the observed value. To calculate the expected value, we generated 1,000 size-matched reshuffled features (same number and same width) and calculated the number of genetic variants falling within each reshuffled random feature. The median number for these 1,000 permutations is the expected value. Significance was assessed by empirical p value of the observed overlap relative to the distribution of expected overlaps from the set of 1,000 random permuted regions.
Data visualization, clustering and general statistical tests
Visualization and clustering of heat maps was performed using Broad’s Morpheus software (https://software.broadinstitute.org/morpheus). For visualization in heat maps, gene expression was Z transformed by row. This matrix was then clustered based on average Euclidean distance between each gene across the 20 samples. Principal component analysis was performed using the prcomp and autoplot R packages [132]. Venn diagrams were generated using BioVenn [133]. Aggregate plots and heat maps for ChIP-seq and DNase-seq datasets were generated using Deeptools v2.3.3 [128]. Boxplots, bar graphs, cumulative distribution plots, and scatterplots were generated using GraphPad Prism (v7.0e). Specific statistical tests are noted as used. Generally, nonparametric tests comparing values (i.e., boxplots) used Mann-Whitney (M-W) tests, while distribution comparisons (i.e., cumulative frequency plots) used Kolmogorov-Smirnov (KS) tests. For graphs, p-values are marked by asterisks, as follows: *, p < 0.05; **, p < 0.01; ***, p < 0.001; and ns (not significant), p > 0.05. Boxplots are graphed to show 1.5*IQR (interquartile range, Tukey) and bars plots show the SEM around the mean per group.
Gene Ontology and functional enrichment analysis
Functional annotation clustering for all indicated gene lists was performed using DAVID v6.8 [134] with high stringency for clustering, but otherwise using default parameters. Gene Ontology enrichment for molecular function was performed using DAVID using only the field “GOTERM_MF_DIRECT” and the output was ranked on the FDR column. Strain-specific, sex-biased TFs were identified from the list of 301 B6-unique or 207 CAST-unique sex specific gene lists. Strain-shared TFs were identified from the combined list of 211 standard and strict common sex-biased genes, which was filtered based on the associated gene ontology terms using DAVID analysis, based on molecular functions related to DNA binding or transcription factor activity (GO:0003677, GO:0043565, or GO:0000981). For shared sex-biased TFs to be considered expressed, a minimum of 2 of the 6 subgroups (male and female B6, CAST, and CD1 liver) had to have a median expression ≥ 1 FPKM. For a strain-specific TF to be considered expressed, the median expression in that strain must be ≥ 1 FPKM for at least one sex.
Supporting information
Data Availability
Data availability RNA-seq data for male and female B6 and CAST mouse liver are available under accession number GSE130913 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE130913). H3K27ac ChIP-seq data for male and female B6 and CAST mouse liver are available under accession number GSE130909 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE130909). DNase-seq data for male and female B6 and CAST mouse liver are available under accession number (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE130912).
Funding Statement
This work was supported in part by NIH grant DK121998 (to DJW). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
References
- 1.Grath S, Parsch J. Sex-Biased Gene Expression. Annual review of genetics. 2016;50:29–44. Epub 2016/08/31. doi: 10.1146/annurev-genet-120215-035429 . [DOI] [PubMed] [Google Scholar]
- 2.Rigby N, Kulathinal RJ. Genetic Architecture of Sexual Dimorphism in Humans. Journal of cellular physiology. 2015;230(10):2304–10. Epub 2015/03/06. doi: 10.1002/jcp.24979 . [DOI] [PubMed] [Google Scholar]
- 3.Parsch J, Ellegren H. The evolutionary causes and consequences of sex-biased gene expression. Nature reviews Genetics. 2013;14(2):83–7. Epub 2013/01/19. doi: 10.1038/nrg3376 . [DOI] [PubMed] [Google Scholar]
- 4.Zheng W, Xu H, Lam SH, Luo H, Karuturi RK, Gong Z. Transcriptomic analyses of sexual dimorphism of the zebrafish liver and the effect of sex hormones. PloS one. 2013;8(1):e53562. Epub 2013/01/26. doi: 10.1371/journal.pone.0053562 ; PubMed Central PMCID: PMC3547925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Qiao Q, Le Manach S, Sotton B, Huet H, Duvernois-Berthet E, Paris A, et al. Deep sexual dimorphism in adult medaka fish liver highlighted by multi-omic approach. Sci Rep. 2016;6:32459. Epub 2016/08/27. doi: 10.1038/srep32459 ; PubMed Central PMCID: PMC5000296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kwekel JC, Desai VG, Moland CL, Branham WS, Fuscoe JC. Age and sex dependent changes in liver gene expression during the life cycle of the rat. BMC Genomics. 2010;11:675. Epub 2010/12/02. doi: 10.1186/1471-2164-11-675 ; PubMed Central PMCID: PMC3012673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wauthier V, Waxman DJ. Sex-specific early growth hormone response genes in rat liver. Molecular endocrinology (Baltimore, Md). 2008;22(8):1962–74. Epub 2008/05/17. doi: 10.1210/me.2007-0549 ; PubMed Central PMCID: PMC2725767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Rinn JL, Rozowsky JS, Laurenzi IJ, Petersen PH, Zou K, Zhong W, et al. Major molecular differences between mammalian sexes are involved in drug metabolism and renal function. Dev Cell. 2004;6(6):791–800. Epub 2004/06/05. doi: 10.1016/j.devcel.2004.05.005 . [DOI] [PubMed] [Google Scholar]
- 9.Clodfelter KH, Holloway MG, Hodor P, Park SH, Ray WJ, Waxman DJ. Sex-dependent liver gene expression is extensive and largely dependent upon signal transducer and activator of transcription 5b (STAT5b): STAT5b-dependent activation of male genes and repression of female genes revealed by microarray analysis. Molecular endocrinology (Baltimore, Md). 2006;20(6):1333–51. Epub 2006/02/14. doi: 10.1210/me.2005-0489 . [DOI] [PubMed] [Google Scholar]
- 10.Lowe R, Gemma C, Rakyan VK, Holland ML. Sexually dimorphic gene expression emerges with embryonic genome activation and is dynamic throughout development. BMC Genomics. 2015;16(1):295. Epub 2015/04/19. doi: 10.1186/s12864-015-1506-4 ; PubMed Central PMCID: PMC4410000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhang Y, Klein K, Sugathan A, Nassery N, Dombkowski A, Zanger UM, et al. Transcriptional profiling of human liver identifies sex-biased genes associated with polygenic dyslipidemia and coronary artery disease. PloS one. 2011;6(8):e23506. Epub 2011/08/23. doi: 10.1371/journal.pone.0023506 ; PubMed Central PMCID: PMC3155567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Etheridge AS, Gallins PJ, Jima D, Broadaway KA, Ratain MJ, Schuetz E, et al. A New Liver Expression Quantitative Trait Locus Map From 1,183 Individuals Provides Evidence for Novel Expression Quantitative Trait Loci of Drug Response, Metabolic, and Sex-Biased Phenotypes. Clinical pharmacology and therapeutics. 2020;107(6):1383–93. Epub 2019/12/24. doi: 10.1002/cpt.1751 ; PubMed Central PMCID: PMC7816646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Lau-Corona D, Suvorov A, Waxman DJ. Feminization of Male Mouse Liver by Persistent Growth Hormone Stimulation: Activation of Sex-Biased Transcriptional Networks and Dynamic Changes in Chromatin States. Mol Cell Biol. 2017;37(19). Epub 2017/07/12. doi: 10.1128/MCB.00301-17 ; PubMed Central PMCID: PMC5599723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Warnefors M, Mössinger K, Halbert J, Studer T, VandeBerg JL, Lindgren I, et al. Sex-biased microRNA expression in mammals and birds reveals underlying regulatory mechanisms and a role in dosage compensation. Genome Res. 2017;27(12):1961–73. Epub 2017/10/29. doi: 10.1101/gr.225391.117 ; PubMed Central PMCID: PMC5741053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hao P, Waxman DJ. Functional Roles of Sex-Biased, Growth Hormone-Regulated MicroRNAs miR-1948 and miR-802 in Young Adult Mouse Liver. Endocrinology. 2018;159(3):1377–92. Epub 2018/01/19. doi: 10.1210/en.2017-03109 ; PubMed Central PMCID: PMC5839735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Melia T, Hao P, Yilmaz F, Waxman DJ. Hepatic Long Intergenic Noncoding RNAs: High Promoter Conservation and Dynamic, Sex-Dependent Transcriptional Regulation by Growth Hormone. Mol Cell Biol. 2016;36(1):50–69. Epub 2015/10/16. doi: 10.1128/MCB.00861-15 ; PubMed Central PMCID: PMC4702595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Melia T, Waxman DJ. Sex-Biased lncRNAs Inversely Correlate With Sex-Opposite Gene Coexpression Networks in Diversity Outbred Mouse Liver. Endocrinology. 2019;160(5):989–1007. Epub 2019/03/07. doi: 10.1210/en.2018-00949 ; PubMed Central PMCID: PMC6449536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Goldfarb CN, Waxman DJ. Global analysis of expression, maturation and subcellular localization of mouse liver transcriptome identifies novel sex-biased and TCPOBOP-responsive long non-coding RNAs. BMC Genomics. 2021;22(1):212. Epub 2021/03/26. doi: 10.1186/s12864-021-07478-5 ; PubMed Central PMCID: PMC7992343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Lorbek G, Perse M, Horvat S, Bjorkhem I, Rozman D. Sex differences in the hepatic cholesterol sensing mechanisms in mice. Molecules. 2013;18(9):11067–85. Epub 2013/09/13. doi: 10.3390/molecules180911067 ; PubMed Central PMCID: PMC6270450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Waxman DJ, Holloway MG. Sex differences in the expression of hepatic drug metabolizing enzymes. Mol Pharmacol. 2009;76(2):215–28. Epub 2009/06/02. doi: 10.1124/mol.109.056705 ; PubMed Central PMCID: PMC2713118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Anderson GD. Chapter 1 Gender Differences in Pharmacological Response. Epilepsy in Women—The Scientific Basis for Clinical Management. International Review of Neurobiology2008. p. 1–10. [Google Scholar]
- 22.Cho J, Kim L, Li Z, Rose NR, Talor MV, Njoku DB. Sex bias in experimental immune-mediated, drug-induced liver injury in BALB/c mice: suggested roles for Tregs, estrogen, and IL-6. PloS one. 2013;8(4):e61186. Epub 2013/04/12. doi: 10.1371/journal.pone.0061186 ; PubMed Central PMCID: PMC3618451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Mennecozzi M, Landesmann B, Palosaari T, Harris G, Whelan M. Sex differences in liver toxicity-do female and male human primary hepatocytes react differently to toxicants in vitro? PloS one. 2015;10(4):e0122786. Epub 2015/04/08. doi: 10.1371/journal.pone.0122786 ; PubMed Central PMCID: PMC4388670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Buzzetti E, Parikh PM, Gerussi A, Tsochatzis E. Gender differences in liver disease and the drug-dose gender gap. Pharmacol Res. 2017;120:97–108. Epub 2017/03/25. doi: 10.1016/j.phrs.2017.03.014 . [DOI] [PubMed] [Google Scholar]
- 25.Della Torre S. Non-alcoholic Fatty Liver Disease as a Canonical Example of Metabolic Inflammatory-Based Liver Disease Showing a Sex-Specific Prevalence: Relevance of Estrogen Signaling. Frontiers in endocrinology. 2020;11:572490. Epub 2020/10/20. doi: 10.3389/fendo.2020.572490 ; PubMed Central PMCID: PMC7531579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Cvitanovic Tomas T, Urlep Z, Moskon M, Mraz M, Rozman D. LiverSex Computational Model: Sexual Aspects in Hepatic Metabolism and Abnormalities. Frontiers in physiology. 2018;9:360. Epub 2018/05/01. doi: 10.3389/fphys.2018.00360 ; PubMed Central PMCID: PMC5907313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Natri HM, Wilson MA, Buetow KH. Distinct molecular etiologies of male and female hepatocellular carcinoma. BMC cancer. 2019;19(1):951. Epub 2019/10/17. doi: 10.1186/s12885-019-6167-2 ; PubMed Central PMCID: PMC6794913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Brie B, Ramirez MC, De Winne C, Lopez Vicchi F, Villarruel L, Sorianello E, et al. Brain Control of Sexually Dimorphic Liver Function and Disease: The Endocrine Connection. Cell Mol Neurobiol. 2019;39(2):169–80. Epub 2019/01/19. doi: 10.1007/s10571-019-00652-0 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Farhy LS, Bowers CY, Veldhuis JD. Model-projected mechanistic bases for sex differences in growth hormone regulation in humans. Am J Physiol Regul Integr Comp Physiol. 2007;292(4):R1577–93. Epub 2006/12/23. doi: 10.1152/ajpregu.00584.2006 . [DOI] [PubMed] [Google Scholar]
- 30.Waters MJ, Brooks AJ. JAK2 activation by growth hormone and other cytokines. The Biochemical journal. 2015;466(1):1–11. Epub 2015/02/07. doi: 10.1042/BJ20141293 ; PubMed Central PMCID: PMC4325515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chia DJ. Minireview: mechanisms of growth hormone-mediated gene regulation. Molecular endocrinology (Baltimore, Md). 2014;28(7):1012–25. Epub 2014/05/16. doi: 10.1210/me.2014-1099 ; PubMed Central PMCID: PMC4075164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Jansson JO, Eden S, Isaksson O. Sexual dimorphism in the control of growth hormone secretion. Endocr Rev. 1985;6(2):128–50. Epub 1985/01/01. doi: 10.1210/edrv-6-2-128 [DOI] [PubMed] [Google Scholar]
- 33.Jaffe CA, Turgeon DK, Lown K, Demott-Friberg R, Watkins PB. Growth hormone secretion pattern is an independent regulator of growth hormone actions in humans. American journal of physiology Endocrinology and metabolism. 2002;283(5):E1008–15. Epub 2002/10/12. doi: 10.1152/ajpendo.00513.2001 . [DOI] [PubMed] [Google Scholar]
- 34.Dhir RN, Dworakowski W, Thangavel C, Shapiro BH. Sexually dimorphic regulation of hepatic isoforms of human cytochrome p450 by growth hormone. The Journal of pharmacology and experimental therapeutics. 2006;316(1):87–94. Epub 2005/09/15. doi: 10.1124/jpet.105.093773 . [DOI] [PubMed] [Google Scholar]
- 35.Wauthier V, Sugathan A, Meyer RD, Dombkowski AA, Waxman DJ. Intrinsic sex differences in the early growth hormone responsiveness of sex-specific genes in mouse liver. Molecular endocrinology (Baltimore, Md). 2010;24(3):667–78. Epub 2010/02/13. doi: 10.1210/me.2009-0454 ; PubMed Central PMCID: PMC2840812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Connerney J, Lau-Corona D, Rampersaud A, Waxman DJ. Activation of Male Liver Chromatin Accessibility and STAT5-Dependent Gene Transcription by Plasma Growth Hormone Pulses. Endocrinology. 2017;158(5):1386–405. Epub 2017/03/23. doi: 10.1210/en.2017-00060 ; PubMed Central PMCID: PMC6283433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Holloway MG, Laz EV, Waxman DJ. Codependence of growth hormone-responsive, sexually dimorphic hepatic gene expression on signal transducer and activator of transcription 5b and hepatic nuclear factor 4alpha. Molecular endocrinology (Baltimore, Md). 2006;20(3):647–60. Epub 2005/10/22. doi: 10.1210/me.2005-0328 . [DOI] [PubMed] [Google Scholar]
- 38.Hao P, Waxman DJ. STAT5 Regulation of Sex-Dependent Hepatic CpG Methylation at Distal Regulatory Elements Mapping to Sex-Biased Genes. Mol Cell Biol. 2021;41(2). Epub 2020/11/18. doi: 10.1128/MCB.00166-20 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Chikada H, Ida K, Ando E, Inagaki Y, Sakamoto A, Kamiya A. Establishment and analysis of a mouse model that regulates sex-related differences in liver drug metabolism. Lab Invest. 2018;98(11):1500–11. Epub 2018/07/04. doi: 10.1038/s41374-018-0088-6 . [DOI] [PubMed] [Google Scholar]
- 40.Zhang Y, Laz EV, Waxman DJ. Dynamic, sex-differential STAT5 and BCL6 binding to sex-biased, growth hormone-regulated genes in adult mouse liver. Mol Cell Biol. 2012;32(4):880–96. Epub 2011/12/14. doi: 10.1128/MCB.06312-11 ; PubMed Central PMCID: PMC3272977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Conforto TL, Zhang Y, Sherman J, Waxman DJ. Impact of CUX2 on the female mouse liver transcriptome: activation of female-biased genes and repression of male-biased genes. Mol Cell Biol. 2012;32(22):4611–27. Epub 2012/09/12. doi: 10.1128/MCB.00886-12 ; PubMed Central PMCID: PMC3486175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Villar D, Berthelot C, Aldridge S, Rayner TF, Lukk M, Pignatelli M, et al. Enhancer evolution across 20 mammalian species. Cell. 2015;160(3):554–66. Epub 2015/01/31. doi: 10.1016/j.cell.2015.01.006 ; PubMed Central PMCID: PMC4313353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Carelli FN, Liechti A, Halbert J, Warnefors M, Kaessmann H. Repurposing of promoters and enhancers during mammalian evolution. Nat Commun. 2018;9(1):4066. Epub 2018/10/06. doi: 10.1038/s41467-018-06544-z ; PubMed Central PMCID: PMC6172195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wong ES, Thybert D, Schmitt BM, Stefflova K, Odom DT, Flicek P. Decoupling of evolutionary changes in transcription factor binding and gene expression in mammals. Genome Res. 2015;25(2):167–78. Epub 2014/11/15. doi: 10.1101/gr.177840.114 ; PubMed Central PMCID: PMC4315291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Stefflova K, Thybert D, Wilson MD, Streeter I, Aleksic J, Karagianni P, et al. Cooperativity and rapid evolution of cobound transcription factors in closely related mammals. Cell. 2013;154(3):530–40. Epub 2013/08/06. doi: 10.1016/j.cell.2013.07.007 ; PubMed Central PMCID: PMC3732390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wong ES, Schmitt BM, Kazachenka A, Thybert D, Redmond A, Connor F, et al. Interplay of cis and trans mechanisms driving transcription factor binding and gene expression evolution. Nat Commun. 2017;8(1):1092. Epub 2017/10/25. doi: 10.1038/s41467-017-01037-x ; PubMed Central PMCID: PMC5653656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Andrikopoulos S, Massa CM, Aston-Mourney K, Funkat A, Fam BC, Hull RL, et al. Differential effect of inbred mouse strain (C57BL/6, DBA/2, 129T2) on insulin secretory function in response to a high fat diet. J Endocrinol. 2005;187(1):45–53. Epub 2005/10/11. doi: 10.1677/joe.1.06333 . [DOI] [PubMed] [Google Scholar]
- 48.Shi Z, Wakil AE, Rockey DC. Strain-specific differences in mouse hepatic wound healing are mediated by divergent T helper cytokine responses. Proc Natl Acad Sci U S A. 1997;94(20):10663–8. Epub 1997/10/06. doi: 10.1073/pnas.94.20.10663 PubMed Central PMCID: PMC23440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Melia T, Waxman DJ. Genetic factors contributing to extensive variability of sex-specific hepatic gene expression in Diversity Outbred mice. PloS one. 2020;15(12):e0242665. Epub 2020/12/03. doi: 10.1371/journal.pone.0242665 ; PubMed Central PMCID: PMC7710091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Wang S, Chen L, Wang Q, He Z, Chen S, Zhang H, et al. Strain differences between CD-1 and C57BL/6 mice in expression of metabolic enzymes and DNA methylation modifications of the primary hepatocytes. Toxicology. 2019;412:19–28. Epub 2018/12/06. doi: 10.1016/j.tox.2018.11.011 . [DOI] [PubMed] [Google Scholar]
- 51.Goncalves A, Leigh-Brown S, Thybert D, Stefflova K, Turro E, Flicek P, et al. Extensive compensatory cis-trans regulation in the evolution of mouse gene expression. Genome Res. 2012;22(12):2376–84. Epub 2012/08/25. doi: 10.1101/gr.142281.112 ; PubMed Central PMCID: PMC3514667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sugathan A, Waxman DJ. Genome-wide analysis of chromatin states reveals distinct mechanisms of sex-dependent gene regulation in male and female mouse liver. Mol Cell Biol. 2013;33(18):3594–610. Epub 2013/07/10. doi: 10.1128/MCB.00280-13 ; PubMed Central PMCID: PMC3753870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lau-Corona D, Bae WK, Hennighausen L, Waxman DJ. Sex-biased genetic programs in liver metabolism and liver fibrosis are controlled by EZH1 and EZH2. PLoS Genet. 2020;16(5):e1008796. Epub 2020/05/20. doi: 10.1371/journal.pgen.1008796 ; PubMed Central PMCID: PMC7263639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Matthews BJ, Waxman DJ. Impact of 3D genome organization, guided by cohesin and CTCF looping, on sex-biased chromatin interactions and gene expression in mouse liver. Epigenetics & chromatin. 2020;13(1):30. Epub 2020/07/19. doi: 10.1186/s13072-020-00350-y ; PubMed Central PMCID: PMC7368777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Reizel Y, Spiro A, Sabag O, Skversky Y, Hecht M, Keshet I, et al. Gender-specific postnatal demethylation and establishment of epigenetic memory. Genes Dev. 2015;29(9):923–33. Epub 2015/05/03. doi: 10.1101/gad.259309.115 ; PubMed Central PMCID: PMC4421981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Abu-Toamih Atamni HJ, Kontogianni G, Binenbaum I, Mott R, Himmelbauer H, Lehrach H, et al. Hepatic gene expression variations in response to high-fat diet-induced impaired glucose tolerance using RNAseq analysis in collaborative cross mouse population. Mammalian genome: official journal of the International Mammalian Genome Society. 2019;30(9–10):260–75. Epub 2019/10/28. doi: 10.1007/s00335-019-09816-1 . [DOI] [PubMed] [Google Scholar]
- 57.Clodfelter KH, Miles GD, Wauthier V, Holloway MG, Zhang X, Hodor P, et al. Role of STAT5a in regulation of sex-specific gene expression in female but not male mouse liver revealed by microarray analysis. Physiological genomics. 2007;31(1):63–74. doi: 10.1152/physiolgenomics.00055.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Krebs CJ, Khan S, MacDonald JW, Sorenson M, Robins DM. Regulator of sex-limitation KRAB zinc finger proteins modulate sex-dependent and -independent liver metabolism. Physiol Genomics. 2009;38(1):16–28. Epub 2009/04/09. doi: 10.1152/physiolgenomics.90391.2008 ; PubMed Central PMCID: PMC2696148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Sueyoshi T, Kobayashi R, Nishio K, Aida K, Moore R, Wada T, et al. A nuclear factor (NF2d9) that binds to the male-specific P450 (Cyp 2d-9) gene in mouse liver. Mol Cell Biol. 1995;15(8):4158–66. Epub 1995/08/01. doi: 10.1128/MCB.15.8.4158 PubMed Central PMCID: PMC230654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Churchill GA, Gatti DM, Munger SC, Svenson KL. The Diversity Outbred mouse population. Mammalian genome: official journal of the International Mammalian Genome Society. 2012;23(9–10):713–8. Epub 2012/08/16. doi: 10.1007/s00335-012-9414-2 ; PubMed Central PMCID: PMC3524832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Keane TM, Goodstadt L, Danecek P, White MA, Wong K, Yalcin B, et al. Mouse genomic variation and its effect on phenotypes and gene regulation. Nature. 2011;477(7364):289–94. Epub 2011/09/17. doi: 10.1038/nature10413 ; PubMed Central PMCID: PMC3276836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Schmidt CW. Diversity outbred: a new generation of mouse model. Environ Health Perspect. 2015;123(3):A64–7. Epub 2015/03/03. doi: 10.1289/ehp.123-A64 ; PubMed Central PMCID: PMC4348734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Consortium GP. A global reference for human genetic variation. Nature. 2015;526(7571):68. doi: 10.1038/nature15393 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.French JE, Gatti DM, Morgan DL, Kissling GE, Shockley KR, Knudsen GA, et al. Diversity Outbred Mice Identify Population-Based Exposure Thresholds and Genetic Factors that Influence Benzene-Induced Genotoxicity. Environ Health Perspect. 2015;123(3):237–45. Epub 2014/11/07. doi: 10.1289/ehp.1408202 ; PubMed Central PMCID: PMC4348743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Svenson KL, Gatti DM, Valdar W, Welsh CE, Cheng R, Chesler EJ, et al. High-resolution genetic mapping using the Mouse Diversity outbred population. Genetics. 2012;190(2):437–47. Epub 2012/02/22. doi: 10.1534/genetics.111.132597 ; PubMed Central PMCID: PMC3276626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Wade CM, Kulbokas EJ 3rd, Kirby AW, Zody MC, Mullikin JC, Lander ES, et al. The mosaic structure of variation in the laboratory mouse genome. Nature. 2002;420(6915):574–8. Epub 2002/12/06. doi: 10.1038/nature01252 . [DOI] [PubMed] [Google Scholar]
- 67.Geraldes A, Basset P, Gibson B, Smith KL, Harr B, Yu HT, et al. Inferring the history of speciation in house mice from autosomal, X-linked, Y-linked and mitochondrial genes. Mol Ecol. 2008;17(24):5349–63. Epub 2009/01/06. doi: 10.1111/j.1365-294X.2008.04005.x ; PubMed Central PMCID: PMC2915842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Aldinger KA, Sokoloff G, Rosenberg DM, Palmer AA, Millen KJ. Genetic variation and population substructure in outbred CD-1 mice: implications for genome-wide association studies. PloS one. 2009;4(3):e4729. Epub 2009/03/07. doi: 10.1371/journal.pone.0004729 ; PubMed Central PMCID: PMC2649211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lin S, Lin Y, Nery JR, Urich MA, Breschi A, Davis CA, et al. Comparison of the transcriptional landscapes between human and mouse tissues. Proceedings of the National Academy of Sciences. 2014;111(48):17224–9. doi: 10.1073/pnas.1413624111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Li Z, Tuteja G, Schug J, Kaestner KH. Foxa1 and Foxa2 are essential for sexual dimorphism in liver cancer. Cell. 2012;148(1–2):72–83. Epub 2012/01/24. doi: 10.1016/j.cell.2011.11.026 ; PubMed Central PMCID: PMC3266536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Laz EV, Holloway MG, Chen CS, Waxman DJ. Characterization of three growth hormone-responsive transcription factors preferentially expressed in adult female liver. Endocrinology. 2007;148(7):3327–37. Epub 2007/04/07. doi: 10.1210/en.2006-1192 ; PubMed Central PMCID: PMC2585771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Takasugi M, Hayakawa K, Arai D, Shiota K. Age- and sex-dependent DNA hypomethylation controlled by growth hormone in mouse liver. Mech Ageing Dev. 2013;134(7–8):331–7. Epub 2013/05/28. doi: 10.1016/j.mad.2013.05.003 . [DOI] [PubMed] [Google Scholar]
- 73.Shen L, Shao NY, Liu X, Maze I, Feng J, Nestler EJ. diffReps: detecting differential chromatin modification sites from ChIP-seq data with biological replicates. PloS one. 2013;8(6):e65598. Epub 2013/06/14. doi: 10.1371/journal.pone.0065598 ; PubMed Central PMCID: PMC3677880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Matthews BJ, Waxman DJ. Computational prediction of CTCF/cohesin-based intra-TAD loops that insulate chromatin contacts and gene expression in mouse liver. Elife. 2018;7. Epub 2018/05/15. doi: 10.7554/eLife.34077 ; PubMed Central PMCID: PMC5986275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Farber CR, Medrano JF. Fine mapping reveals sex bias in quantitative trait loci affecting growth, skeletal size and obesity-related traits on mouse chromosomes 2 and 11. Genetics. 2007;175(1):349–60. Epub 2006/11/18. doi: 10.1534/genetics.106.063693 ; PubMed Central PMCID: PMC1775020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Garieri M, Delaneau O, Santoni F, Fish RJ, Mull D, Carninci P, et al. The effect of genetic variation on promoter usage and enhancer activity. Nat Commun. 2017;8(1):1358. Epub 2017/11/09. doi: 10.1038/s41467-017-01467-7 ; PubMed Central PMCID: PMC5677018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Maurano MT, Haugen E, Sandstrom R, Vierstra J, Shafer A, Kaul R, et al. Large-scale identification of sequence variants influencing human transcription factor occupancy in vivo. Nat Genet. 2015;47(12):1393–401. Epub 2015/10/27. doi: 10.1038/ng.3432 ; PubMed Central PMCID: PMC4666772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Reddy TE, Gertz J, Pauli F, Kucera KS, Varley KE, Newberry KM, et al. Effects of sequence variation on differential allelic transcription factor occupancy and gene expression. Genome Res. 2012;22(5):860–9. Epub 2012/02/04. doi: 10.1101/gr.131201.111 ; PubMed Central PMCID: PMC3337432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Conforto TL, Steinhardt GFt, Waxman DJ. Cross Talk Between GH-Regulated Transcription Factors HNF6 and CUX2 in Adult Mouse Liver. Molecular endocrinology (Baltimore, Md). 2015;29(9):1286–302. Epub 2015/07/29. doi: 10.1210/me.2015-1028 ; PubMed Central PMCID: PMC4552435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Meyer RD, Laz EV, Su T, Waxman DJ. Male-specific hepatic Bcl6: growth hormone-induced block of transcription elongation in females and binding to target genes inversely coordinated with STAT5. Molecular endocrinology (Baltimore, Md). 2009;23(11):1914–26. Epub 2009/10/03. doi: 10.1210/me.2009-0242 ; PubMed Central PMCID: PMC2775936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Gil N, Ulitsky I. Regulation of gene expression by cis-acting long non-coding RNAs. Nature reviews Genetics. 2020;21(2):102–17. Epub 2019/11/16. doi: 10.1038/s41576-019-0184-5 . [DOI] [PubMed] [Google Scholar]
- 82.Tong C, Yin Y. Localization of RNAs in the nucleus: cis- and trans- regulation. RNA Biol. 2021:1–14. Epub 2021/03/09. doi: 10.1080/15476286.2021.1894025 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Bordon Y. TOX for tired T cells. Nature reviews Immunology. 2019;19(8):476. Epub 2019/06/28. doi: 10.1038/s41577-019-0193-9 . [DOI] [PubMed] [Google Scholar]
- 84.Wang X, He Q, Shen H, Xia A, Tian W, Yu W, et al. TOX promotes the exhaustion of antitumor CD8(+) T cells by preventing PD1 degradation in hepatocellular carcinoma. Journal of hepatology. 2019;71(4):731–41. Epub 2019/06/08. doi: 10.1016/j.jhep.2019.05.015 . [DOI] [PubMed] [Google Scholar]
- 85.Wolf G, Yang P, Fuchtbauer AC, Fuchtbauer EM, Silva AM, Park C, et al. The KRAB zinc finger protein ZFP809 is required to initiate epigenetic silencing of endogenous retroviruses. Genes Dev. 2015;29(5):538–54. Epub 2015/03/05. doi: 10.1101/gad.252767.114 ; PubMed Central PMCID: PMC4358406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Coskran TM, Jiang Z, Klaunig JE, Mager DL, Obert L, Robertson A, et al. Induction of endogenous retroelements as a potential mechanism for mouse-specific drug-induced carcinogenicity. PloS one. 2017;12(5):e0176768. Epub 2017/05/05. doi: 10.1371/journal.pone.0176768 ; PubMed Central PMCID: PMC5417610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Jiang S, Minter LC, Stratton SA, Yang P, Abbas HA, Akdemir ZC, et al. TRIM24 suppresses development of spontaneous hepatic lipid accumulation and hepatocellular carcinoma in mice. Journal of hepatology. 2015;62(2):371–9. Epub 2014/10/05. doi: 10.1016/j.jhep.2014.09.026 ; PubMed Central PMCID: PMC4772153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Herquel B, Ouararhni K, Khetchoumian K, Ignat M, Teletin M, Mark M, et al. Transcription cofactors TRIM24, TRIM28, and TRIM33 associate to form regulatory complexes that suppress murine hepatocellular carcinoma. Proceedings of the National Academy of Sciences. 2011;108(20):8212–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Hanna D, Sugamori KS, Bott D, Grant DM. The impact of sex on hepatotoxic, inflammatory and proliferative responses in mouse models of liver carcinogenesis. Toxicology. 2020;442:152546. Epub 2020/08/09. doi: 10.1016/j.tox.2020.152546 . [DOI] [PubMed] [Google Scholar]
- 90.Lewis L, Chappell GA, Kobets T, O’Brian BE, Sangaraju D, Kosyk O, et al. Sex-specific differences in genotoxic and epigenetic effects of 1,3-butadiene among mouse tissues. Archives of toxicology. 2019;93(3):791–800. Epub 2018/12/16. doi: 10.1007/s00204-018-2374-x ; PubMed Central PMCID: PMC6451682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Israel JW, Chappell GA, Simon JM, Pott S, Safi A, Lewis L, et al. Tissue- and strain-specific effects of a genotoxic carcinogen 1,3-butadiene on chromatin and transcription. Mammalian genome: official journal of the International Mammalian Genome Society. 2018;29(1–2):153–67. Epub 2018/02/13. doi: 10.1007/s00335-018-9739-6 ; PubMed Central PMCID: PMC6095468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Maronpot RR. Biological basis of differential susceptibility to hepatocarcinogenesis among mouse strains. Journal of toxicologic pathology. 2009;22(1):11–33. doi: 10.1293/tox.22.11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Rogers AB. Stress of Strains: Inbred Mice in Liver Research. Gene Expr. 2018;19(1):61–7. Epub 2018/08/11. doi: 10.3727/105221618X15337408678723 ; PubMed Central PMCID: PMC6290319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Mansour MR, Abraham BJ, Anders L, Berezovskaya A, Gutierrez A, Durbin AD, et al. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element. Science. 2014;346(6215):1373–7. doi: 10.1126/science.1259037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Carey M, Lin YS, Green MR, Ptashne M. A mechanism for synergistic activation of a mammalian gene by GAL4 derivatives. Nature. 1990;345(6273):361–4. Epub 1990/05/24. doi: 10.1038/345361a0 [DOI] [PubMed] [Google Scholar]
- 96.Dukler N, Gulko B, Huang Y-F, Siepel A. Is a super-enhancer greater than the sum of its parts? Nature Genetics. 2016;49:2. doi: 10.1038/ng.3759 https://www.nature.com/articles/ng.3759#supplementary-information. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Shin HY, Willi M, Yoo KH, Zeng X, Wang C, Metser G, et al. Hierarchy within the mammary STAT5-driven Wap super-enhancer. Nature genetics. 2016;48(8):904. doi: 10.1038/ng.3606 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Carleton JB, Berrett KC, Gertz J. Multiplex Enhancer Interference Reveals Collaborative Control of Gene Regulation by Estrogen Receptor alpha-Bound Enhancers. Cell Syst. 2017;5(4):333–44 e5. Epub 2017/10/02. doi: 10.1016/j.cels.2017.08.011 ; PubMed Central PMCID: PMC5679353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Huang J, Li K, Cai W, Liu X, Zhang Y, Orkin SH, et al. Dissecting super-enhancer hierarchy based on chromatin interactions. Nat Commun. 2018;9(1):943. Epub 2018/03/07. doi: 10.1038/s41467-018-03279-9 ; PubMed Central PMCID: PMC5838163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hay D, Hughes JR, Babbs C, Davies JOJ, Graham BJ, Hanssen L, et al. Genetic dissection of the alpha-globin super-enhancer in vivo. Nat Genet. 2016;48(8):895–903. Epub 2016/07/05. doi: 10.1038/ng.3605 ; PubMed Central PMCID: PMC5058437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Moorthy SD, Davidson S, Shchuka VM, Singh G, Malek-Gilani N, Langroudi L, et al. Enhancers and super-enhancers have an equivalent regulatory role in embryonic stem cells through regulation of single or multiple genes. Genome research. 2017;27(2):246–58. doi: 10.1101/gr.210930.116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lee HK, Willi M, Shin HY, Liu C, Hennighausen L. Progressing super-enhancer landscape during mammary differentiation controls tissue-specific gene regulation. Nucleic Acids Res. 2018;46(20):10796–809. Epub 2018/10/05. doi: 10.1093/nar/gky891 ; PubMed Central PMCID: PMC6237736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Levo M, Zalckvar E, Sharon E, Machado ACD, Kalma Y, Lotam-Pompan M, et al. Unraveling determinants of transcription factor binding outside the core binding site. Genome research. 2015;25(7):1018–29. doi: 10.1101/gr.185033.114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Jolma A, Yin Y, Nitta KR, Dave K, Popov A, Taipale M, et al. DNA-dependent formation of transcription factor pairs alters their binding specificity. Nature. 2015;527(7578):384. doi: 10.1038/nature15518 [DOI] [PubMed] [Google Scholar]
- 105.Faure AJ, Schmidt D, Watt S, Schwalie PC, Wilson MD, Xu H, et al. Cohesin regulates tissue-specific expression by stabilizing highly occupied cis-regulatory modules. Genome Res. 2012;22(11):2163–75. Epub 2012/07/12. doi: 10.1101/gr.136507.111 ; PubMed Central PMCID: PMC3483546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Duan J, Shi J, Ge X, Dolken L, Moy W, He D, et al. Genome-wide survey of interindividual differences of RNA stability in human lymphoblastoid cell lines. Sci Rep. 2013;3:1318. Epub 2013/02/21. doi: 10.1038/srep01318 ; PubMed Central PMCID: PMC3576867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Tang Z, Luo OJ, Li X, Zheng M, Zhu JJ, Szalaj P, et al. CTCF-Mediated Human 3D Genome Architecture Reveals Chromatin Topology for Transcription. Cell. 2015;163(7):1611–27. Epub 2015/12/22. doi: 10.1016/j.cell.2015.11.024 ; PubMed Central PMCID: PMC4734140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Hnisz D, Weintraub AS, Day DS, Valton AL, Bak RO, Li CH, et al. Activation of proto-oncogenes by disruption of chromosome neighborhoods. Science. 2016;351(6280):1454–8. Epub 2016/03/05. doi: 10.1126/science.aad9024 ; PubMed Central PMCID: PMC4884612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Lupianez DG, Kraft K, Heinrich V, Krawitz P, Brancati F, Klopocki E, et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell. 2015;161(5):1012–25. Epub 2015/05/12. doi: 10.1016/j.cell.2015.04.004 ; PubMed Central PMCID: PMC4791538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Dixon JR, Selvaraj S, Yue F, Kim A, Li Y, Shen Y, et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature. 2012;485(7398):376–80. Epub 2012/04/13. doi: 10.1038/nature11082 ; PubMed Central PMCID: PMC3356448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Hnisz D, Day DS, Young RA. Insulated Neighborhoods: Structural and Functional Units of Mammalian Gene Control. Cell. 2016;167(5):1188–200. Epub 2016/11/20. doi: 10.1016/j.cell.2016.10.024 ; PubMed Central PMCID: PMC5125522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Odhams CA, Cortini A, Chen L, Roberts AL, Vinuela A, Buil A, et al. Mapping eQTLs with RNA-seq reveals novel susceptibility genes, non-coding RNAs and alternative-splicing events in systemic lupus erythematosus. Hum Mol Genet. 2017;26(5):1003–17. Epub 2017/01/08. doi: 10.1093/hmg/ddw417 ; PubMed Central PMCID: PMC5409091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Tan JY, Smith AAT, Ferreira da Silva M, Matthey-Doret C, Rueedi R, Sonmez R, et al. cis-Acting Complex-Trait-Associated lincRNA Expression Correlates with Modulation of Chromosomal Architecture. Cell Rep. 2017;18(9):2280–8. Epub 2017/03/02. doi: 10.1016/j.celrep.2017.02.009 . [DOI] [PubMed] [Google Scholar]
- 114.Waszak SM, Delaneau O, Gschwind AR, Kilpinen H, Raghav SK, Witwicki RM, et al. Population Variation and Genetic Control of Modular Chromatin Architecture in Humans. Cell. 2015;162(5):1039–50. Epub 2015/08/25. doi: 10.1016/j.cell.2015.08.001 . [DOI] [PubMed] [Google Scholar]
- 115.Gaffney DJ. Global properties and functional complexity of human gene regulatory variation. PLoS Genet. 2013;9(5):e1003501. Epub 2013/06/06. doi: 10.1371/journal.pgen.1003501 ; PubMed Central PMCID: PMC3667745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Dejager L, Libert C, Montagutelli X. Thirty years of Mus spretus: a promising future. Trends Genet. 2009;25(5):234–41. Epub 2009/04/14. doi: 10.1016/j.tig.2009.03.007 . [DOI] [PubMed] [Google Scholar]
- 117.Percie du Sert N, Hurst V, Ahluwalia A, Alam S, Avey MT, Baker M, et al. The ARRIVE guidelines 2.0: Updated guidelines for reporting animal research. PLoS Biol. 2020;18(7):e3000410. Epub 2020/07/15. doi: 10.1371/journal.pbio.3000410 ; PubMed Central PMCID: PMC7360023 following competing interests: AA is the editor in chief of the British Journal of Pharmacology. WJB, ICC, and ME are authors of the original ARRIVE guidelines. WJB serves on the Independent Statistical Standing Committee of the funder CHDI foundation. AC is a Senior Editor for PLOS ONE. AC, CJM, MM, and ESS were involved in the IICARus trial. ME, MM, and ESS have received funding from NC3Rs. ME sits on the MRC ERPIC panel. STH is chair of the NC3Rs board; trusteeship of the BLF, Kennedy Trust, DSRU, and CRUK; member of Governing Board, Nuffield Council of Bioethics, member Science Panel for Health (EU H2020); founder and NEB Director Synairgen; consultant Novartis, Teva, and AZ; and chair MRC/GSK EMINENT Collaboration. VH, KL, EJP, and NPdS are NC3Rs staff; role includes promoting the ARRIVE guidelines. SEL and UD are on the advisory board of the UK Reproducibility Network. CJM has shareholdings in Hindawi, is on the publishing board of the Royal Society, and on the EU Open Science policy platform. UD, MM, NPdS, CJM, ESS, TS, and HW are members of EQIPD. MM is a member of the Animals in Science Committee and on the steering group of the UK Reproducibility Network. NPdS and TS are associate editors of BMJ Open Science. OHP is vice president of Academia Europaea, editor in chief of Function, senior executive editor of the Journal of Physiology, and member of the Board of the European Commission’s SAPEA (Science Advice for Policy by European Academies). FR is an NC3Rs board member and has shareholdings in GSK. FR and NAK have shareholdings in AstraZeneca. PR is a member of the University of Florida Institutional Animal Care and Use Committee and editorial board member of Shock. ESS is editor in chief of BMJ Open Science. SDS’s role is to provide expertise and does not represent the opinion of the NIH. TS has shareholdings in Johnson & Johnson. SA, MTA, MB, PG, DWH, and KR declared no conflict of interest. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Storch KF, Lipan O, Leykin I, Viswanathan N, Davis FC, Wong WH, et al. Extensive and divergent circadian gene expression in liver and heart. Nature. 2002;417(6884):78–83. Epub 2002/04/23. doi: 10.1038/nature744 . [DOI] [PubMed] [Google Scholar]
- 119.Ling G, Sugathan A, Mazor T, Fraenkel E, Waxman DJ. Unbiased, genome-wide in vivo mapping of transcriptional regulatory elements reveals sex differences in chromatin structure associated with sex-specific liver gene expression. Mol Cell Biol. 2010;30(23):5531–44. Epub 2010/09/30. doi: 10.1128/MCB.00601-10 ; PubMed Central PMCID: PMC2976433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Lodato NJ, Rampersaud A, Waxman DJ. Impact of CAR Agonist Ligand TCPOBOP on Mouse Liver Chromatin Accessibility. Toxicol Sci. 2018;164(1):115–28. Epub 2018/04/05. doi: 10.1093/toxsci/kfy070 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Schmidt D, Wilson MD, Spyrou C, Brown GD, Hadfield J, Odom DT. ChIP-seq: using high-throughput sequencing to discover protein-DNA interactions. Methods. 2009;48(3):240–8. Epub 2009/03/12. doi: 10.1016/j.ymeth.2009.03.001 ; PubMed Central PMCID: PMC4052679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 2009;25(9):1105–11. Epub 2009/03/18. doi: 10.1093/bioinformatics/btp120 ; PubMed Central PMCID: PMC2672628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Lodato NJ, Melia T, Rampersaud A, Waxman DJ. Sex-Differential Responses of Tumor Promotion-Associated Genes and Dysregulation of Novel Long Noncoding RNAs in Constitutive Androstane Receptor-Activated Mouse Liver. Toxicological sciences: an official journal of the Society of Toxicology. 2017; 159(1):25–41. Epub 2017/09/15. doi: 10.1093/toxsci/kfx114 ; PubMed Central PMCID: PMC5837275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30(7):923–30. Epub 2013/11/15. doi: 10.1093/bioinformatics/btt656 . [DOI] [PubMed] [Google Scholar]
- 125.Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40. Epub 2009/11/17. doi: 10.1093/bioinformatics/btp616 ; PubMed Central PMCID: PMC2796818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9(4):357–9. Epub 2012/03/06. doi: 10.1038/nmeth.1923 ; PubMed Central PMCID: PMC3322381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 2008;9(9):R137. Epub 2008/09/19. doi: 10.1186/gb-2008-9-9-r137 ; PubMed Central PMCID: PMC2592715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ramirez F, Ryan DP, Gruning B, Bhardwaj V, Kilpert F, Richter AS, et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 2016;44(W1):W160–5. Epub 2016/04/16. doi: 10.1093/nar/gkw257 ; PubMed Central PMCID: PMC4987876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Vietri Rudan M, Barrington C, Henderson S, Ernst C, Odom DT, Tanay A, et al. Comparative Hi-C reveals that CTCF underlies evolution of chromosomal domain architecture. Cell Rep. 2015;10(8):1297–309. Epub 2015/03/04. doi: 10.1016/j.celrep.2015.02.004 ; PubMed Central PMCID: PMC4542312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Broman KW, Gatti DM, Simecek P, Furlotte NA, Prins P, Sen Ś, et al. R/qtl2: Software for Mapping Quantitative Trait Loci with High-Dimensional Data and Multiparent Populations. Genetics. 2019;211(2):495–502. Epub 2018/12/29. doi: 10.1534/genetics.118.301595 ; PubMed Central PMCID: PMC6366910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Arnold P, Erb I, Pachkov M, Molina N, van Nimwegen E. MotEvo: integrated Bayesian probabilistic methods for inferring regulatory sites and motifs on multiple alignments of DNA sequences. Bioinformatics. 2011;28(4):487–94. [DOI] [PubMed] [Google Scholar]
- 132.Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5(10):R80. Epub 2004/10/06. doi: 10.1186/gb-2004-5-10-r80 ; PubMed Central PMCID: PMC545600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Hulsen T, de Vlieg J, Alkema W. BioVenn—a web application for the comparison and visualization of biological lists using area-proportional Venn diagrams. BMC Genomics. 2008;9:488. Epub 2008/10/18. doi: 10.1186/1471-2164-9-488 ; PubMed Central PMCID: PMC2584113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44–57. Epub 2009/01/10. doi: 10.1038/nprot.2008.211 . [DOI] [PubMed] [Google Scholar]