Skip to main content

A primer on the use of mouse models for identifying direct sex chromosome effects that cause sex differences in non-gonadal tissues

Abstract

In animals with heteromorphic sex chromosomes, all sex differences originate from the sex chromosomes, which are the only factors that are consistently different in male and female zygotes. In mammals, the imbalance in Y gene expression, specifically the presence vs. absence of Sry, initiates the differentiation of testes in males, setting up lifelong sex differences in the level of gonadal hormones, which in turn cause many sex differences in the phenotype of non-gonadal tissues. The inherent imbalance in the expression of X and Y genes, or in the epigenetic impact of X and Y chromosomes, also has the potential to contribute directly to the sexual differentiation of non-gonadal cells. Here, we review the research strategies to identify the X and Y genes or chromosomal regions that cause direct, sexually differentiating effects on non-gonadal cells. Some mouse models are useful for separating the effects of sex chromosomes from those of gonadal hormones. Once direct “sex chromosome effects” are detected in these models, further studies are required to narrow down the list of candidate X and/or Y genes and then to identify the sexually differentiating genes themselves. Logical approaches to the search for these genes are reviewed here.

Background

In animals with an unmatched (heteromorphic) pair of sex chromosomes, all sex differences in the phenotype originate from the unequal effects of the sex chromosomes because they are the only factors that consistently differ between male and female zygotes. In species such as mammals in which the sex chromosomes are XX and XY, a fundamental goal is to identify the sex-biasing effects of the two sex chromosomes on phenotypes. This review discusses the strategies for identifying X and Y genes or mechanisms that cause sexual bias, using mouse models that differ in their sex chromosome complement, including differences in the parental source of the X chromosome.

Primary among the sexually unbalanced effects of X and Y genes are those that cause sexual differentiation of the gonads. In mammals, the most important sex differences in the gonads are thought to be initiated by the Y-linked gene Sry, which is expressed in the indifferent gonadal ridge of males and induces there a cascade of molecular and cellular events that commit that tissue to a testicular fate [14]. In the absence of Sry in the XX female, ovaries develop through an active developmental program that includes inhibition of testicular pathways [5]. Therefore, the developmentally limited male-specific expression of Sry in the gonads sets up a lifelong sex difference in secretion of testicular vs. ovarian hormones. Because sex steroid hormones, especially androgens and estrogens, are considered the major class of proximate signals that act throughout the body to cause sexual differentiation, the Sry effect has long been thought to be primarily responsible for determining the sexual phenotype of most mammalian species.

In addition to the effects of Sry on gonadal tissues, X and Y genes may be expressed at different levels in XX and XY non-gonadal cells as a consequence of their imbalance in the genomes of all XX and XY cells. By virtue of this imbalance, X and Y genes are known to cause numerous sex differences in phenotype [611]. These X and Y gene effects are here called “direct” effects of the sex chromosomes, because the sex bias in the X and Y gene expressions acts directly to cause sex differences in non-gonadal tissues instead of indirectly via an action on the gonads to induce sex differences caused by gonadal hormones [12]. Historically, it has been much easier to study the effects of gonadal hormones, which can be administered or blocked or withdrawn by simple experimental procedures. It has been much more difficult to discover the direct sex-biased effects of the X and Y genes on non-gonadal tissues, because altering the sex chromosome complement usually causes changes in the type or function of the gonads, and therefore introduces confounding differences in the level of gonadal secretions.

Here, we review mouse models (see also [13]) that enable the separation of sex chromosome complement effects from the hormonal confounds, thus facilitating the identification of direct sex chromosome effects (direct SCEs) that contribute to sex differences. We focus especially on direct SCEs that are expected to contribute to sex differences in the function of normal XX and XY cells. The goal of our studies, and of this review, is not only to discuss how to detect direct SCEs but also to map out a strategy for identifying the X or Y genes that cause sex differences in cells. The strategy represents a logic tree, which progressively narrows down the potential candidate genes responsible for direct SCEs.

Anatomy of the mouse sex chromosomes and possible causes of sex differences in phenotype

The mammalian sex chromosomes are thought to have evolved from an ancestral pair of autosomes. One autosome, the proto-Y chromosome, acquired a dominant male-determining locus, which led to the loss of recombination with the proto-X, and ultimately, to the wholesale loss of gene-encoding DNA from the Y chromosome [14, 15]. However, there have been gene additions to the X and/or Y chromosome subsequent to their divergence [1620]. The present-day Y chromosome is usually small and gene-poor relative to the X chromosome [21, 22]. However, the mouse Y has an unusually high gene count as a result of massive gene amplification that is thought to be driven by an ongoing post-meiotic X-Y genomic conflict [22, 23]. During meiosis, the X and Y chromosomes pair at the pseudoautosomal regions (PARs), thus enabling the X and Y PARs to recombine [2427]. The X and Y PARs are therefore identical on average between males and females and are not thought to cause sex differences in phenotypes. The number and type of sex chromosomes present in females or males are referred to as the sex chromosome complement, and we use this term to encompass the parental imprinting and inactivation status of the X chromosomes (see below). For the purposes of this review, we break down the sex chromosome complement into a number of components that could contribute to sex differences in phenotypes. Aside from the PARs, which are present in two doses in female and male mice, the genomic dosage of these components is not the same in males and females (Table 1 and Fig. 1). Thus, females have two copies of the non-PAR region of the X chromosome (NPX), whereas males have one NPX and one non-PAR Y (NPY). Females also differ from males in that they receive an X chromosome from each parent, whereas males only receive an X from their mother. Because of parental imprinting, gene expression can differ between the maternal and paternal X chromosomes. Also, as is discussed more fully below, one of the X chromosomes in females is “inactivated.” Thus, about half of female cells inactivate the maternal X chromosome and experience the paternal X imprint (unlike any male cell), whereas the other half of the cells inactivate the paternal X chromosome and experience the maternal X imprint (similar to male cells). The inherent imbalance in the sex chromosome complement that results in a difference in gene expression between XX and XY cells can therefore be summarized as follows: (1) the absence vs. the presence of the NPY genes, (2) the difference in genomic dose of NPX genes (two vs. one copies), (3) any differences due to the parental imprint on the NPX genes, and (4) the presence vs. the absence of an inactive X chromosome (Table 1).

Table 1 XX vs. XY genomic dose for sex chromosome complement components
Fig. 1
figure 1

Major parts of the mouse X and Y chromosomes. Top: The 170-Mb X chromosome is divided into the pseudoautosomal region (PAR), containing the Sts gene and a few others, and the non-PAR region (NPX). The 89-Mb Y chromosome comprises the PAR and non-PAR Y (NPY). The Sry gene is on the Y chromosome short arm. Bottom: XX progeny inherit X chromosomes from the mother (Xm) and father (Xp). The XX mouse is a mosaic of cells expressing one of the parental X chromosomes. Green shows the patches of tissues in which Xp is active, and lavender shows the patches in which Xm is active. The XY mouse inherits the Xm and expresses only that X chromosome in all tissues. CEN centromere

Without any compensatory mechanism, the double genomic representation of X genes in XX cells, compared to the single representation of those genes in XY cells, would create an imbalance in X gene expression between the sexes (F>M); such a chromosome-wide sexual imbalance in expression is thought to be disadvantageous because X genes would not be in balance in both sexes with autosomal genes that they drive or that drive them [28]. X-inactivation, the process by which one X chromosome is largely transcriptionally silenced in non-germline XX cells, is an effective mechanism that reduces the imbalance in X gene expression. However, X-inactivation does not eliminate sex differences in the expression of X genes but reduces it to match approximately the level of sexual dimorphism in the expression of genes encoded on autosomes, which are present in two copies in each sex [29]. It also introduces the possibility of epigenetic effects of the inactive X, which are discussed more fully below.

X inactivation leads to mosaicism of X gene expression in XX mice. In eutherian mammals such as the mouse, the choice to inactivate the paternal or maternal X chromosome is random and patchy in tissues stemming from the inner cell mass of the blastocyst (i.e., the tissues giving rise to the body of the embryo), so that some somatic XX cells predominantly express X genes inherited from the mother, and others express X genes inherited from the father [30]. In outbred or natural populations that carry genetic polymorphisms, there are two types of X mosaicism—of the alleles and imprints. Because the maternally derived X chromosome will have many alleles that are different from those on the paternally derived X chromosome, roughly half of the cells in the XX mice will express the paternal X alleles and show the effects of any paternal imprint on X genes, and the other half will express the maternal X alleles and evince the effects of any maternal imprint on X genes. Thus, females are expected to have fewer extreme tissue phenotypes, because the extreme effects of specific X alleles (e.g., causing abnormal function) is often buffered by the expression of another allele in other cells of the same individual [3133]. These effects are thought to lead to sex differences in the variability of tissue phenotypes (XX females less variable than XY males), and broader adaptiveness of female tissues to diverse environments because of the inherently greater diversity of X alleles within an individual female vs. an individual male. However, an overall reduced variability of X gene expression in human females, relative to males, has not been supported [34] but might be important under extreme environmental conditions or disease.

Thus, we can expand the list of possible sources of sex differences in phenotype arising from differences in sex chromosome complement, to include the following: (5) The inherent mosaicism of XX tissues arising from random X inactivation, which can lead to less phenotypic variability among females than males and to average differences in phenotype between the sexes; (6) sex differences in the prevalence of specific X alleles that may also lead to average phenotypic differences between the sexes. However, when studying inbred mice, all X alleles are the same in the two sexes, so these additional sources of sex differences are not operative.

Finally, it is possible that the difference in sex chromosome complement between normal males and females can lead to direct SCEs that are linked to the epigenetic status of the sex chromosomes rather than differences in X and Y gene expression [3538]. One example of this arises from the fact that in somatic cells, the inactive X (Xi) in females is rich in heterochromatin, reflecting the limited transcriptional activity. Heterochromatinization depends on enzymes that introduce repressive marks on histones, and it is hypothesized that heterochromatinization of the X competes with the introduction of repressive marks at some autosomal loci, thus affecting autosomal gene expression in females. This could be linked to a direct SCE even though it is not linked to an XX vs. XY difference in X gene expression. We do not consider this type of epigenetic contribution to direct SCEs further.

Two mouse models that identify direct SCEs

An ideal mouse model for identifying direct SCEs is one that provides mice that differ in their sex chromosome complement (XX vs. XY) but not in the level of gonadal secretions. In practice, this is an elusive goal, but some experimental paradigms are informative even if strict control of gonadal hormone levels may not always be possible.

Sf1 KO mice

One approach to eliminating the hormonal confound is to study mice that lack gonads. The most useful gonadless mice to date are those lacking steroidogenic factor 1 (Sf1, also known as Nr5a1 and Ad4bp). Sf1-null mice of both sexes are born without gonads or adrenals and have a malformed hypothalamus [39, 40]. They die at birth because of the lack of glucocorticoids but are kept alive by injections of corticosterone and transplantation of adrenals [41, 42]. Such gonadless mice show XX vs. XY differences in some neural phenotypes, indicating that some sex differences, seen in gonadally intact mice in the same litters, are caused by direct non-gonadal effects arising from the difference in sex chromosome complement [43]. This model is valuable because it compares XX and XY mice or tissues that have never been exposed to gonadal secretions. However, the model has two main disadvantages: the homozygous Sf1-null mice are produced from heterozygous null parents, so that each litter produces a relatively small number of homozygous null pups, and each homozygous mouse must receive an adrenal transplant. We do not consider this model further.

“Four core genotypes” (FCG) mice

The FCG model [44], in which the Sry gene causing testis development has been “moved” from the Y chromosome to an autosome, is so far the model most often used for separating the effects of sex chromosome complement from the effects of gonadal type. In general, the model can be used to identify sex differences caused by gonadal hormones (testicular vs. ovarian secretions) independently of sex chromosome complement (aside from Sry) and to identify sex differences caused by the direct effects of sex chromosome complement on non-gonadal tissues. The essence of this model is that the Y chromosome (here designated Yˉ) lacks the small region encoding Sry, and a functional Sry transgene has been introduced onto an autosome. XYˉ mice possessing the Sry transgene (XYˉSry mice) have testes and are fertile (XYM). Because Sry now segregates independently of the sex chromosomes, crossing XYM with normal XX female mice yields the four core genotypes: XX mice with ovaries (XXF) or testes (XXM), and XY mice with ovaries (XYF) or testes (XYM). Comparison of the FCG progeny is a 2 × 2 comparison of individuals that are either XX or XYˉ and have or lack the Sry transgene (Table 2).

Table 2 Progeny of FCG Cross XX x XYˉSry (XYM)

At the outset, it is important to establish that the XX vs. XY phenotypic sex difference of interest is seen in the FCG XXF vs. XYM comparison. Comparing the phenotype of mice with or without Sry is a measure of the effects of the Sry transgene: both the direct effects of Sry on tissues themselves and the indirect effects caused by testicular vs. ovarian secretions. Comparing XX and XYˉ mice allows assessment of the differential effects of sex chromosome complement (XX vs. XYˉ). Importantly, the comparison of XX and XYˉ in this model identifies differences that are common to the XXF vs. XYF and XXM vs. XYM comparisons and excludes differences found in normal XX females vs. XY males that are due to the Sry expression in males. The FCG does not separate the potential direct effects of Sry outside of the gonads from the indirect effects that are a consequence of Sry triggering testicular development, because the direct effects and hormonal effects are confounded (Table 2).

Although the FCG cross has been used successfully to identify a number of direct SCEs [12, 45, 46], investigators should be aware of the caveats considered in the next section.

Important caveats relating to the FCG cross

Genetic background

FCG mice have been generated on various genetic backgrounds—C57BL/6J inbred [47, 48], SJL inbred [49], C3H/He x C57BL/6 hybrid with a fixed B6 X [38], and “MF1” outbred with a fixed (“uniform”) MF1 X [50] (see “Genetic variability of NPX” section). However, with the C57BL/6J inbred FCG stock now available from the Jackson Laboratory (strain 010905) and difficulties with availability and maintenance of the F1 and outbred strains, the Jackson Laboratory strain is becoming the strain of choice.

Potential differences in expression between endogenous and transgenic Sry

The key features of the FCG cross are (i) the deletion of Sry from the Y (this Yˉ, originally derived from the strain 129/SvEv-Gpi1c) and (ii) the Sry transgene [44]. The Sry transgene is located on chromosome 3 and is estimated to be present in 12–14 copies, although they may not all be functional [51]. Thus, the transgene may not have expression levels that are identical to those of the endogenous Sry encoded by the Y chromosome [52]; furthermore, there may be some ectopic expression relative to the endogenous Sry. If the differences between XX females and XY males depend on whether the Sry gene is wild-type (WT) or transgenic (as in FCG males), then the FCG model could misrepresent the normal differences in WT XX vs. XY cells. However, the XX vs. XYˉ difference in the FCG model is assessed both in the presence and absence of the transgenic Sry, so that XX vs. XYˉ differences that occur under both conditions are likely to be robust and independent of Sry effects. To avoid the possibility of any abnormal effects of the transgene, some studies compare only XX and XYˉ females [53]. Furthermore, elements of the XX vs. XYˉ differences found in FCG mice (for example, the effects of one vs. two X chromosomes) can be potentially confirmed using the XY* model that has no transgene (see section on “Linking direct SCEs to a specific component(s) of the XX or XY complements” below) [47, 48].

Genetic variability of NPX

When comparing the FCG genotypes, the NPX segments in XX and XY mice should be genetically identical, so that group differences attributed to sex chromosome complement (XX vs. XYˉ) are not confounded by NPX allelic differences. In inbred strains such as C57BL/6 (“B6”) or SJL, all the Xs are identical so the requirement for genetic uniformity of NPX is met.

Differences in hormonal status in groups with the same type of gonad

Another issue for the FCG model is that the two groups of gonadal females and the two groups of gonadal males differ markedly in their fertility, which could be linked to hormonal differences. XYF on a B6 background are almost always sterile (PSB unpublished data), although on the MF1 outbred background, they do breed [54, 55]. Using the MF1 strain, it has been shown that it is a combination of a markedly reduced oocyte pool together with the expression of the Y-encoded transcription factor ZFY2 in the oocytes leading to impaired development of embryos prior to implantation, which severely limits the fertility [55]. Nevertheless, estrous cycle data collected in association with an FCG behavioral study [56] revealed that estrous cycles are still present in ~85–90% of MF1 XYF at 6–8 months of age (William Davies, personal communication), demonstrating that the majority of XYF ovaries are hormonally competent. FCG XYF on the B6 background have also recently been shown to cycle when assayed at 35–65 days of age and have similar levels of estradiol to XXF. However, ovaries are smaller in XYF than in XXF, and gonadotrophin levels are elevated in XYF relative to XXF especially after day 65, suggesting ovarian dysfunction or premature failure in XYF relative to XXF [9].

XXM are sterile with very small testes. This is because the presence of two X chromosomes in the germ cells leads to early spermatogenic failure [57, 58]. Nevertheless, XXM testes secrete androgens and the levels in XXM and XYM have been reported to be equivalent in adults [9, 52, 59, 60]; furthermore, numerous traits sensitive to testosterone levels during development are similar in XXM and XYM [44, 51, 61, 62]. For example, the anogenital distance is the same in XXM and XYM mice, and smaller in XXF and XYF relative to males, suggesting that the prenatal androgen levels that masculinize anogenital distance are similar in the XXM and XYM [51]. XXM are also reported to have elevated levels of follicle-stimulating hormone (FSH) relative to XYM from before puberty to age 5 months [9]. However, numerous studies have compared levels of testosterone in adult XXM and XYM, or of estradiol in adult XXF and XYF, and none has uncovered differences in plasma gonadal steroid levels to date [9, 52, 59, 60, 6365].

Despite evidence against XX vs. XY differences in levels of circulating gonadal hormones, it is impossible to rule out differences that might occur before birth or in environmental conditions (stress, disease) when hormone levels are not measured. What steps can be taken to avoid the potential confound between these hormone differences and sex chromosome complement differences? One approach that has been used is to gonadectomize the mice (with or without equivalent hormone replacement) so that the phenotype of interest can be measured in mice that have the same levels of gonadal hormones at the time of measurement. However, although this approach controls for acute (“activational”) effects of hormones, it does not control for long term (“organizational”) effects of hormones prior to the gonadectomy [66]. Nevertheless, the FCG model allows an independent assessment of the importance of organizational effects of gonadal hormones, because the comparison of gonadectomized males and females tests for the importance of permanent effects of testicular vs. ovarian secretions. In some cases, robust differences between gonadectomized XX and XY mice have been found in the absence of evidence of organizational hormonal effects [47, 48, 67, 68], so that it is difficult to attribute those XX vs. XY differences to within-sex differences in the levels of gonadal hormones. Investigators may wish to measure phenotypes of gonadally intact FCG mice to test the effects of sex chromosome complement in the presence of ovaries or testes but are encouraged to test gonadectomized mice as well (with or without treatment with equal levels of gonadal hormone at the time of testing) to attempt to control hormone levels experimentally to better understand their role (see “FCG outcomes” section).

FCG outcomes

In what follows, we define sex difference to be a difference in phenotype between WT XX females and WT XY males that has been confirmed to be present in the FCG XXF vs. XYM comparison. The four outcomes correspond to outcomes of a 2 × 2 ANOVA with factors of Sry (absent or present, same as gonadal female vs. male) and sex chromosome complement (XX vs. XY) (Fig. 2).

Fig. 2
figure 2

Possible outcomes of experiments using the four core genotypes to assess the effects of sex chromosome complement (XX vs. XYˉ) and gonadal sex (males M vs. females F) on a dependent variable (DV)

  1. 1.

    An XX vs. XYˉ difference in phenotype occurs in both gonadal females and gonadal males (Fig. 2a), which is equivalent to the original sex difference found in WT mice. This outcome in the ANOVA is a main effect of sex chromosome complement in the absence of a main effect of Sry or an interaction. This result suggests that the sex difference is due to a direct SCE that is independent of Sry/gonadal secretions. Although this conclusion is satisfactory by itself, further tests are prudent, both to confirm the direct SCE in a different model (e.g., in XY* progeny, see next section on “Linking direct SCEs to a specific component(s) of the XX or XY complements”) or to test further for effects of gonadal hormones to rule them out. For example, it is possible to gonadectomize mice with or without hormone replacement, to rule out subtle hormonal confounds. Sex differences are rarely totally unaffected by gonadal hormones.

  2. 2.

    A phenotypic difference is found between mice differing in Sry/gonadal type, which is irrespective of sex chromosome complement and equivalent to the original sex difference found in WT mice. In the two-way ANOVA, this is a main effect of Sry/gonadal type in the absence of effects of sex chromosome complement (Fig. 2b). This result implies that the original sex difference is exclusively an effect of gonadal type (testicular vs. ovarian secretions) and/or a direct effect of Sry acting outside the gonads. Gonadectomy of FCG mice will confirm whether or not there is hormonal involvement and can help differentiate between activational and organizational hormone effects [67, 69].

  3. 3.

    The sex difference is independently influenced by Sry/gonadal type and by a direct SCE. This result leads to a significant main effect of both factors in the two-way ANOVA without a significant interaction (Fig. 2c). To investigate the hormones involved, and their times of action, one would perform further studies to manipulate the levels of gonadal hormones in both sexes either in adulthood or during early phases of development. A number of organizational effects are known to occur postnatally and thus can be revealed by gonadectomy, and prenatal organizational effects may be documented by manipulating hormone levels in utero.

  4. 4.

    The sex difference is caused by an interaction of sex chromosome complement and Sry/gonadal type, as is shown by a statistically significant interaction of the two factors in the two-way ANOVA (Fig. 2d) [47]. This result means that both factors contribute to the sex difference but that the effect of each factor depends on the level of the other factor (e.g., the effect of testicular secretions depends on whether the mouse is XX or XYˉ). Further experiments (gonadectomy, hormone treatments, etc.) would define the hormones involved and their times and sites of action in XX vs. XYˉ mice, and mechanistic experiments would define the molecular pathways influenced by both factors.

Linking direct SCEs to a specific component(s) of the XX or XY complements

The strategies outlined below are based on the assumption that the direct SCEs are a consequence of differences in gene expression resulting from the difference in sex chromosome complement. If the tissue underlying the direct SCE can be identified with a high degree of confidence, then a case may be made for using RNA sequencing (RNAseq) on this tissue from the FCG in order to provide a list of candidate genes at this stage (see sections below beginning with “Candidate genes for direct SCEs). We will refer to this tissue as the “target tissue.” The candidate genes will be either on the NPX or NPY. The following crosses provide a logical approach to determining the extent to which NPX and/or NPY genes are responsible for direct SCEs.

Important: see Additional file 1, which provides the details of the three-generation breeding strategy needed to produce the fathers for Cross B. This involves progeny from Cross C.

Identifying NPX and NPY effects using Cross A: XX♀ x XY*♂

This cross is the best starting point for identifying potential NPX and NPY effects. The fathers have a variant Y chromosome (denoted Y*) that leads to the generation of a minute X chromosome derivative (termed Y*X for historical reasons) composed of a complete PAR, no NPY, and a small subsegment of NPX (NPX+) currently estimated to be less than 1% of the total NPX (Table 3, Fig. 3) [70, 71]. As we shall see, the resulting XmY*X female progeny are used for Cross C that enables NPX dosage effects to be distinguished from NPX parental imprinting effects.

Table 3 Progeny of Cross A: XX x XY*
Fig. 3
figure 3

Sex chromosome structure diagrams related to Cross A (XY* model [70]). Sections of the sex chromosomes have been deleted to allow expansion of other regions for illustration. For the WT sex chromosomes, the PAR is illustrated in three sections, one of which contains Sts. The minute + subregion of the NPX (<1% of the total NPX), adjacent to the PAR, is shown as a bracketed region within the entire NPX for the X and XXY* chromosomes, and in yellow for the Y* and Y*X chromosomes. The Y* chromosome has an X centromere and the plus (+) region of NPX adjacent to a unique duplicated PAR that nevertheless lacks Sts (ΔPAR). The X centromere (X CEN) is the functional centromere, whereas the Y centromere is inactive (Y CEN). The Y* chromosome undergoes abnormal recombination with the X chromosome to produce (1) the long XY* chromosome that is an end-to-end fusion of NPX and NPY with the ΔPAR lacking Sts and (2) the minute Y*X chromosome with a normal PAR and the plus (+) region of NPX. The gene map for the Y*X illustrates the approximate positions of genes transcribed from the forward strand (black rectangles) or reverse strand (lines), with the centromere (orange) and the plus (+) region of NPX (yellow) color coded. In effect, Y*X is an X chromosome with a massive NPX deletion running from a region thought to be just proximal to Msl3 and ending within the Sstx/Spin2 multi-copy cluster adjacent to the telomeric X centromere [22, 72, 118120]. The dividing line between NPX+ and the rest of NPX is not known precisely but is on the centromeric side of Msl3 [72]. This figure updates and corrects earlier versions [50, 121].

The PAR sections AC were originally defined by mapping using the XY* model [71]: A, the terminal section of Mid1/Fxy (including the last three exons) that lies within the PAR [122]; B, the region that contains the multi-copy locus DXYHgu1 [123]; and C, the region containing Sts [124]. It is now known that Erdr1 is located in the PAR just distal to Mid1 and is retained in Y* and XY*—thus, it maps to the PAR region B [125]. On the other hand, Asmt is absent in Y* and XY*—thus, it maps to the PAR region C [126]. Sts is thought to be distal to Asmt, but this has not been confirmed

  1. 1.

    Identifying an effect of NPX (two vs. one copies).

    A comparison of XmXp vs. XmY*X is a test of the effect of the number of NPX segments (2 vs. 1), keeping the number of PARs = 2.

    Cross A also tests for an NPX effect in the comparison of the two gonadal male groups, XmXpY* with XmY*, although in this case, the comparison deviates from the precise genetic conditions found in the XX-XY comparison in three ways: the presence of NPX+ in XmY*; single copy of Sts in XmY* instead of two copies found in XX or XY; and three copies of the region of the PAR that does not contain Sts, instead of two in XX or XY (Table 3). If the phenotypic difference between females containing one vs. two copies of NPX is similar to the difference between males with one vs. two copies of NPX, these deviations from normal XX and XY are generally considered minor [47].

  2. 2.

    Identifying an NPY effect.

    The effect of NPY can be detected in a comparison of XmY*X vs. XmY* and XmXp vs. XmXpY*. These comparisons are confounded with differences in the number of PAR segments as shown in Table 3. The Y* also has a different strain origin (LT/Sv) from that of the Yˉ from the FCG (strain 129/SvEv-Gpi1c). Thus, if the investigator finds no effect of the Y chromosome in this cross, it could be a Y chromosome strain difference. On the other hand, if a Y effect is found, then one can conclude that at least some Y chromosomes create a difference relative to XY*X mice, which therefore may explain the original sex chromosome effect in FCG. Because the Y* chromosome encodes Sry, the NPY effect could be the result of testicular secretions; that possibility will already have been evaluated by a study of FCG mice, which will indicate whether the phenotype is influenced by gonadal hormones or not. If Cross A is used without knowledge of the outcomes from studies of FCG mice, it will not be possible to eliminate the hormonal confound of the Y* chromosome.

  3. 3.

    Interpretation of outcomes

    1. (a)

      NPX copy number effect only. The different effects of one vs. two copies of NPX can be caused by dosage differences (inherently higher expression of genes that escape X inactivation in XX than XY) or by X imprinting effects (different expression levels of paternally vs. maternally imprinted X chromosomes). Importantly, this cross tests for an effect of NPX copy number both in gonadal males (with Sry) and gonadal females (without Sry). If an NPX copy number effect is found in Cross A and in FCG mice, the results indicate that the effect does not depend on the Sry transgene in the FCG model.

    2. (b)

      NPY effect only. The SCE is attributed to NPY genes. Numerous NPY genes have an X “partner” gene that theoretically has a similar function (see section on “Candidate genes for direct SCEs” below). If the NPY effect is caused by one of these genes, the implication is that the NPY gene has an effect distinct from its X partner.

    3. (c)

      NPX and NPY effects. In this case, it is possible that X and Y partner genes are involved, with the X gene escaping X inactivation [50]. Alternatively or additionally, the NPX effect could be due to imprinting.

  4. 4.

    Advantages and drawbacks

    1. (a)

      This a relatively simple cross that enables comparisons to be made quickly and economically.

    2. (b)

      Some of the XmY*X female offspring can be utilized as mothers for Cross C that enables parental X imprinting effects to be identified.

    3. (c)

      It has the minor disadvantage that the Y* chromosome has a different strain origin to that in the FCG cross.

    4. (c)

      It has the disadvantage that Y* encodes Sry, and thus all NPY-bearing mice are gonadal males, and differ hormonally from gonadal females that lack a Y chromosome. Thus, it does not test for an effect of NPY independent of its effect on gonadal secretions, unless prior studies of FCG mice indicate that the phenotype is not sensitive to gonadal hormones. This disadvantage is remedied in Cross B.

    5. (d)

      NPX is subject to X inactivation in XmXp. A minor concern is that in XmY*X the small NPX+ segment present on Y*X is exempt from X inactivation because it does not include Xist. Genes in this segment are therefore expected to be expressed from both the X and Y*X chromosomes [72]. In contrast, the same segment in XmXp is expected to undergo random X inactivation and thus be expressed only from the active X chromosome. If these genes are expressed in the target tissue of the direct SCE, it is therefore likely that they will be expressed higher in XY*X than XX. However, only a few known genes are encoded by NPX+ (Fig. 3) and they can be discounted if these genes show no difference in expression in the relevant target tissue.

Identifying NPX and NPY effects using Cross B: XX♀ x XYˉY*X Sry

As we have seen, there are a number of drawbacks to Cross A when it is used to attribute direct SCEs to NPX or NPY. This is particularly the case when the FCG outcomes indicate that gonadal hormones affect the direct SCE [50]. Cross B provides a more sophisticated assessment of potential NPX and NPY effects by bringing together the Yˉ chromosome and Sry transgene from the FCG cross, with the Y*X chromosome generated by Cross A. Cross B is useful for balancing the number of PARs when assessing the effect of NPX dose, for example, in the comparison XX vs. XY*X. The cross produces the seven genotypes shown in Table 4, each with or without the Sry transgene, thus 14 genotypes in all. However, it is the three female/male pairs of genotypes with two sex chromosomes that are of primary interest. (The fathers for this cross are generated by mating XmXpY*X females from Cross C to XYˉSry males—see Additional file 1).

Table 4 Progeny of Cross B: XX x XYˉY*X Sry

Cross B allows the following comparisons or interpretations:

  1. 1.

    Confirming the sex chromosome complement effect identified in the FCG cross

    The offspring of this cross include all the genotypes from the FCG cross, so the outcomes relating to XmXp (F or M) vs. XmYˉ (F or M) should be replicated here.

  2. 2.

    Identifying an effect of NPX (two vs. one copies)

    A comparison of XmXp vs. XmY*X is a within-sex test (i.e., the comparison can be made either in gonadal females or in gonadal males) of the effect of the number of NPX segments (two vs. one), keeping the number of PARs = 2.

  3. 3.

    Identifying an NPY [−Sry] effect.

    A comparison of XmY*X vs. XmYˉ provides a within-sex test of the effect of NPY [−Sry], keeping the number of PARs = 2.

  4. 4.

    Interpretation of outcomes

    1. (a)

      NPX copy number effect only. The different effects of one vs. two copies of NPX can be caused by dosage differences (inherently higher expression of genes that escape X inactivation in XX than XY), or by X imprinting effects, as for Cross A. Importantly, this cross tests for an effect of NPX copy number both in gonadal males (with Sry) and in gonadal females (without Sry).

    2. (b)

      NPY [−Sry] effect only. The SCE is attributed to NPY genes. Many NPY genes have an X partner gene that theoretically has a similar function (see section on “Candidate genes for direct SCEs” below). If the NPY effect is caused by one of these genes, the implication is that the NPY gene has an effect distinct from its X partner.

    3. (c)

      NPX and NPY effects. In this case, it is possible that X and Y partner genes are involved, with the X gene escaping X inactivation [50]. Alternatively or additionally, the NPX effect could be due to imprinting.

  5. 5.

    Advantages and drawbacks

    1. (a)

      The fathers for this cross can be bred by mating XmXpY*X females from Cross C to XYˉSry (FCG) males (Additional file 1).

    2. (b)

      This cross allows an elegant comparison of mice with two X chromosomes, one X chromosome, or one X and one Y chromosome (all with two PARs), each of which are produced as males or females. It is the only cross to achieve this degree of balancing of sex chromosome complement and gonadal sex. It also allows more comparisons than any other cross, to assess effects of NPX number, NPY, and gonadal sex.

    3. (c)

      Unlike Cross A, in this cross, NPY effects are not confounded by gonadal sex, and thus hormonal and NPY effects are more easily separated. NPY effects can be detected that occur in both gonadal males and gonadal females.

    4. (d)

      With 14 possible genotypes, the breeding is time-consuming and expensive.

    5. (e)

      Differentiating among the genotypes is difficult using chromosome spreads because the Y*X is minute and easily missed; quantitative genomic PCR can confirm the presence or absence of Y*X [73, 74] (Additional file 2).

    6. (f)

      As for Cross A, a minor concern when detecting an NPX effect is that the small NPX+ segment in XmY*X is present on both sex chromosomes but does not undergo X inactivation, in contrast to the same segment in XmXp that is expected to undergo random X inactivation. If the NPX+ genes are expressed in the target tissue of the direct SCE, it is likely that they will be expressed higher in XY*X than in XX. Similarly, when detecting an NPY effect, the absence vs. presence of NPY [−Sry] is confounded with the presence vs. absence of NPX+ (Fig. 3). However, NPX+ encodes only a small number of genes (Fig. 3) and they can be discounted if these genes show no difference in the expression in the relevant target tissue.

Detecting effects of X chromosome imprinting

An NPX copy number effect identified by Cross A or B can be due to X genes that are exempt from the transcriptional silencing of one X due to random X inactivation [28]. In this case, transcript levels for these X genes are higher in XmXp than in XmY. An NPX copy number effect can also be due to parental X imprinting (see section on “Candidate genes for direct SCEs” below). For some X genes, this results in higher transcription from the Xp and thus elevated transcription in XmXp relative to XmY (an expression pattern similar to that for X genes that escape X inactivation); for other imprinted X genes, it results in higher transcription from the Xm and thus elevated transcription in XmY relative to XmXp. However, in either case, Xm/Xp transcript ratios in the XmXp females can vary from one tissue sample to another if there is patchiness of random X inactivation in the target tissue sample (as exemplified in [30]). Here, we compare crosses that can identify effects arising from X imprinting and that compare female XmY*X (Cross A, Table 3) vs. XpY*X (Cross C, Table 5) in order to avoid the inherent variability of Xm vs. Xp expression associated with random X inactivation in XmXp.

Table 5 Progeny of Cross C: XY*X x XY

An effect of imprinting on X genes is detected by comparing female offspring of Cross A (XX x XY*) with those of Cross C (XY* x XY). As we have seen (Table 3), Cross A generates XmY*X females with a maternally imprinted X, together with XmXp female controls. In Cross C (Table 5), the XmY*X females from Cross A are mated to XY males to produce XpY*X females with a paternally imprinted X, together with XmXp female controls. For reasons that do not concern us here, Cross C also generates some XmXpY*X females and XmYY*X males (Table 5). The important difference in Cross C relative to Cross A is that the XY*X progeny carry a paternal X rather than a maternal X. This comparison is of mice that differ genetically only in the imprint on the single X chromosome.

  1. 1.

    Detecting X p vs. X m imprinting effects

    If an NPX effect detected in Cross A is at least in part caused by a parental imprint of the X chromosome, then a similar or enhanced difference should be detected when comparing XmY*X from Cross A with XpY*X from Cross C.

  2. 2.

    Checking for maternal effects

    Because these differences in imprint are potentially confounded with maternal effects (differences between crosses in maternal behavior, uterine effects, or other environmental differences), it is prudent to compare the XmXp female progeny between the two crosses (A vs. C) as a check for confounding variables. If the XX females from the two crosses are similar, the confounding variables would seem not to be having a significant effect.

  3. 3.

    Advantages and drawbacks

    1. (a)

      These crosses have the advantage that they are both feasible on a B6 inbred background [75, 76]. As previously observed [75], from Cross C, there are no surviving XpOs, but ~11% of the offspring are XpY*X (Rhonda R. Voskuhl, personal communication).

    2. (b)

      Differentiating among the genotypes can be difficult using chromosome spreads because the Y*X is minute and easily missed; quantitative genomic PCR can confirm the presence or absence of Y*X [73, 74].

    3. (c)

      There is evidence from the study of embryos from XO females that maternal X monosomy has deleterious effects on development of preimplantation embryos [55, 77]; these effects could come into play in Cross C. See Additional file 3 for crosses E and F that avoid this potential confound.

    4. (d)

      XpO embryos are developmentally retarded in early pregnancy whereas XmO embryos are not [10, 75, 7779]. This Xp effect would be expected to affect XpY*X embryos and thus is a potential confound when identifying imprinting effects. However, it appears to be ameliorated by the addition of Y*X.

    5. (e)

      The yield of XpY*x progeny in Cross C in B6 is low because of small litters. Probably because of the XpO death in utero, the XmY*X mothers often experience birthing problems leading to the death of viable XpY*X fetuses. A potential solution might be to set up timed B6 XmY*X x XY matings and timed F1 or outbred strain matings known to provide reliable foster mothers. Caesarian section and fostering can then be done when the B6 mothers are due to litter. There are various well-established humane husbandry protocols for this fostering procedure.

Summary: a pragmatic approach

Taking into account the advantages and drawbacks discussed above, the most feasible strategy to investigate sex chromosome complement effects is as follows: (1) Establish a sex difference in gonadally intact XX females vs. XY males. The difference is most likely caused by the effects of gonadal hormones. (2) If experiments such as those recommended by Becker et al. [68] suggest that the sex difference is not caused entirely by differences in levels of gonadal hormones, one can use the FCG mice to detect sex chromosome complement effects that depend on, or are independent of, gonadal hormones. (3) If a sex chromosome complement effect is found in FCG mice, then Crosses A and/or B can be used to provide information as to whether the effect is caused by differences in NPX complement and/or the presence vs. absence of NPY. Cross A is easier than Cross B. Cross B is a more sophisticated cross for identifying NPX and NPY effects and is required to detect NPY effects if FCG outcomes make it difficult to separate hormonal and sex chromosome effects with Cross A. (4) Studies using Crosses A and C will assess the importance of X imprinting vs. NPX dosage. Together, these approaches will narrow down the X or Y candidate genes to be tested further as discussed in the next few sections.

Candidate genes for direct SCEs

Here, we provide an overview of the sex chromosome genes whose differential expression between XX females and XY males may lead to direct SCEs in the target tissue in which the direct SCE originates. These comprise the following: (i) X genes that have parental imprints that lead to differential expression between males and females in the target tissue; (ii) X genes that escape X inactivation in the target tissue, an important group of which are X genes that have functionally similar partner genes on the Y chromosome (X-Y gene pairs that are termed “gametologs”); (iii) Y genes that are expressed in the target tissue of XY males.

Parentally imprinted X genes

Classically, parentally imprinted genes were defined as those that were expressed only from the paternally derived chromosome or from the maternally derived chromosome. Recently, it has been established that parental imprinting can also lead to different levels of expression between the two parentally imprinted chromosomes. These two types of imprinting are now referred to as “canonical” and “noncanonical” imprinting, respectively [80]. There are two examples of canonical X gene imprinting that are relevant here. Firstly, there is the canonical imprinting of the Xist locus (reviewed by [32, 81]). This results in Xist only being expressed from the paternal X of XX embryos, beginning at the two-cell stage, which leads to the progressive inactivation of the paternal X during early cleavage stages. This paternal X inactivation is then removed from the blastocyst inner cell mass (the cells that form the body of the embryo) and is replaced with random Xist-mediated X inactivation, whereas the extra-embryonic tissues that contribute to the placenta retain paternal X inactivation [82, 83]. These Xist imprinting effects have to be borne in mind as a potential confounding factor. For example, paternal X imprinting leads to an early developmental retardation of XpO embryos, which might impact subsequent development [10, 78, 79, 84]. However, this retarding effect of Xp imprinting is much reduced in the context XmXp female vs. XmY male (or female) comparisons [8]. The only other canonically imprinted X genes that have been identified in mice are members of the Xlr gene family that are expressed from the maternal X; mice with only a paternal X exhibit impaired cognitive behavior [80, 8587].

On the other hand, the study of Bonthuis et al. [80] identified 198 noncanonically imprinted X genes with 170 of such genes identified in a single tissue, the hypothalamic arcuate nucleus. In contrast to canonically imprinted genes, the imprinting was very tissue-specific. Clearly, this class of genes has the potential to make a significant contribution to X chromosome-associated direct SCEs. Intriguingly, there is a significant preponderance of X genes showing higher expression from the maternal X across all four tissues analyzed—see Figure S5A and D of Bonthuis et al. [80]. The impact of noncanonical imprinting on SCEs will increase as the expression from the two parental alleles becomes more disparate.

X inactivation escapees

During the evolution of the X-Y chromosomes from their autosomal progenitors, there was a progressive loss of Y genes, while their X partners became subject to X inactivation to balance gene expression between males and females. However, a number of Y genes escaped attrition and the survivors are predominantly widely expressed regulatory genes. It is assumed that dosage sensitivity for these genes led to the retention of the Y copies because mutation of the Y gene was strongly deleterious so that that mutant Y chromosome was not passed on. Retention of the Y gene was associated with escape from inactivation of the X copies, thus retaining two expressing copies in males (X + Y) and females (X + X), preventing a deleterious level of expression relative to interacting autosomal genes [19, 20]. (See Table 6 for the X-Y gene pairs in mice and humans).

Table 6 Mouse and human Y genes and X gametologs

An important caveat is that the set of surviving Y genes varies among different groups of mammals. In mice, there seems to have been a tendency for the X copy of such gene pairs to become subject to X inactivation, and for the Y copy to diverge in sequence and acquire testis-specific functions (Tables 6 and 7). Also, the Y copies of the four X-Y gene pairs where the X is not subject to X inactivation are not essential in mice since XmO mice are viable; indeed, during fetal development, they are slightly ahead of XX fetuses and equivalent to XY fetuses [10]. This is in marked contrast to the lethality of non-mosaic human XOs [88]; this may reflect the larger number (14) of X inactivation escapees among X-Y gene pairs for which human XOs are dosage deficient [19]. Consequently, in mice, there are only four X partner genes that are clear X inactivation escapees as compared to 14 in humans.

Table 7 Mouse ancestral Y genes

Aside from these four X inactivation escapees that have Y partner genes, a number of other X genes show some degree of escape from X inactivation in vivo, and this escape can be tissue specific. The most recent study identified 34 of such genes, giving a total of 38 escapees—approximately 7% of mouse X genes [89].

NPY genes

Because NPY genes are only expressed in males, any NPY gene could potentially contribute to a direct SCE that is identified using the FCG. A comprehensive compendium of the mouse Y gene content has recently been published [22]. Because FCG analyses often seek to shed light on human sex differences, in Table 7, we present the mouse Y gene complement alongside a subset of the human Y gene complement. The omitted human Y genes are those that have been “acquired” by the human Y but not by the mouse Y—for the most part, expression of these genes is restricted to spermatogenic cells [19, 20]. Similarly, the mouse has acquired genes that are not represented on the human Y, and the expression is thought to be restricted to spermatogenic cells; as such, they are unlikely to be involved in direct SCEs. Some of these genes are present in multiple copies and have multiple copy X gametologs. These genes are thought to have been amplified as a consequence of a post-meiotic X vs. Y genomic conflict [22, 23, 90]. The remainders of the mouse and human Y gene complements are termed “ancestral,” because they comprise a group of genes that were present on the Y of therian mammals at an early stage of Y chromosome evolution; all of these genes had gametologs on the X chromosome. Over time, some of the X and Y copies diverged, and in other cases, Y genes were lost differentially among mammalian groups. The mouse Y ancestral gene complement now comprises nine distinct genes, one of which (Zfy) has duplicated and one of which (Rbmy) is estimated to have 30 copies; the human Y has retained 17 ancestral genes, seven of which are also on the mouse Y. The nine ancestral mouse Y genes and the predicted characteristics of the proteins they encode are listed in Table 7.

Identifying candidate genes that underlie direct SCEs

Key information for determining which genes might underlie a direct SCE is their expression profile. RNA sequencing (RNAseq) is the current method of choice for documenting the transcriptome in specific tissues or cell types. It has the potential to document the relative abundance of all the transcripts present in a tissue sample—the detection of rare transcripts can be improved by increasing the sequencing coverage depth. While RNAseq data for the relevant target tissue may be available for XY female vs. XY male comparisons, hormone effects may modulate or nullify expression differences linked to the direct SCE. RNAseq data from FCG mice would help to uncover such effects. In the following sections, we outline how such transcriptome information can be utilized to home in on the X and/or Y genes that generate the direct SCE.

Identifying NPX genes that cause direct SCEs

Here, we consider strategies for identifying the genes underlying a direct SCE when NPX (but not NPY) has been implicated in Cross A or B. The first step is to use the FCG RNAseq data to generate a list of X genes that are differentially expressed in the target tissues of XXF vs. XYF and/or XXM vs. XYM FCG mice, under conditions in which the differences are unlikely to be caused by gonadal hormones.

As we have seen, the most abundant may well be X genes subject to non-canonical parental imprinting effects. If such genes are involved in generating the direct SCE, this should have been detected in the XmY*X vs. XpYX (Cross A vs. C) comparison in the section above entitled “Linking direct SCEs to a specific component(s) of the XX or XY complements.

  1. (a)

    X gene transcript levels where X m Y > X m X p (Table 8). If the canonically imprinted Xlr genes are involved, they will fall into this category. Other X genes falling in this category are prime candidates for being X genes with noncanonical imprinting that leads to reduced expression of Xp relative to Xm. This is the predominant form of noncanonical X gene imprinting (see Figure S5A and D in [80]).

    Table 8 Predicted expression of imprinted X genes
  2. (b)

    X gene transcript levels where X m X p> X m Y (Table 8). This could be due to (i) the noncanonical imprinting where there is greater expression of Xp relative to Xm or (ii) to genes that escape X inactivation, in particular, the four widely expressed mouse X inactivation escapees Kdm5c/Smcx, Kdm6a/Utx, Ddx3x, and Eif2s3x (Table 6) or the additional escapees identified by in vivo studies [89, 91]. The X escapees are among the genes most often found to differ in XX vs. XY mice in microarray-based transcriptome profiling on various adult tissues of FCG mice (Arnold, unpublished), and these have been repeatedly reported to be expressed higher in XX than in XY mice [47, 48, 72, 9297].

The X genes that exhibit expression differences compatible with the direct SCE identified in the FCG mice would be prioritized for further study. The X genes might first be ranked with respect to the fold difference in the level of expression between the relevant FCG genotypes (e.g., XXF vs. XYF), and/or by the p value of a statistical test that estimates the reliability of the difference in the expression between groups. These candidate genes could be further prioritized based on their known characteristics and potential relevance to the phenotypes under investigation. If necessary, candidate genes would be checked using quantitative RT-PCR to confirm the expected group differences. If the candidate NPX gene is a reported X escapee, and is expressed higher in XX than in XY, as expected from its status as an escapee, its importance as a gene causal to the SCE would be established by comparing mice with one vs. two copies of the gene. For example, if Ddx3x (Table 6) is the candidate, one might compare WT XX mice (with two copies of Ddx3x) with XX mice with a heterozygous knockout of Ddx3x (one copy). If these groups of mice differ in the phenotype showing the SCE, such that one vs. two copies of Ddx3x mimics the phenotypic effects of one vs. two copies of NPX, then Ddx3x is a likely contributor to the SCE. This experiment determines if dosage differences in Ddx3x alone are necessary to show the SCE. An additional strategy is to add a transgene encoding Ddx3x to an XO or XY*X mouse, to determine if the phenotype is affected appropriately in mice with one copy vs. more than one copy of the candidate X escapee. The transgenic add-in experiment aims to test if the dosage differences in the X escapee are sufficient by themselves to mimic the SCE. The main caveat is that expression levels of the transgene may be difficult to control depending on its promoter, position effects, etc.

The experiments comparing Cross A vs. C may suggest that the NPX effect is the result of imprinting. In this case, transcriptome profiling may reveal either XX > XY or XY > XX expression patterns (Table 8) and differences in expression of transcripts in XmY*X vs. XpY*X. To provide further strong support for an NPX imprinting explanation of an SCE, one might measure the effects of parent of origin on the epigenetic status of the candidate gene and manipulate its expression to mimic the differences in expression found in XmY*X vs. XpY*X, to demonstrate that these differences in expression alone are necessary and sufficient to mimic the SCE, at least in part.

The manipulation of NPX expression is best performed in a tissue-specific manner, assuming that the tissue of interest is known. For example, a floxed allele of the candidate gene can be knocked down with a tissue-specific Cre. Indeed, the use of tissue-specific knockdown or overexpression may help determine the tissue in which the candidate gene causes the SCE.

Identifying NPY genes that cause direct SCEs

Here, we consider strategies for identifying the genes underlying a direct SCE when NPY (but not NPX) has been implicated in Cross A or C. The first step is to check the RNAseq data to provide a list of the Y genes expressed in the SCE target tissue. Prioritizing the candidate genes may be possible based on the current information about the function of the Y genes or their similar X gametologs.

Given that only seven Y genes (Uba1y, Kdm5d, Uty, Usp9y, Ddx3y, Eif2s3y, and Ssty2) have been shown to be widely expressed in non-gonadal tissues as assayed by RNAseq [22], the “prioritized list” of genes expressed in a target tissue with a putative NPY-linked SCE will likely be limited to one or a very few genes. Thus, it makes sense to move on to the stage of manipulating candidate Y gene expression in order to demonstrate that the SCE of interest is a consequence of the presence vs. the absence of a particular NPY gene or genes. The basic approach is the same as for candidate NPX genes discussed in the last section “Identifying NPX genes that cause direct SCEs”. Keeping other factors constant (e.g., sex chromosomes and hormones), the investigator varies the presence/absence of one candidate Y gene to determine if that manipulation causes a phenotypic difference similar to the SCE caused by the comparison of XX vs. XY. Two basic approaches are available, either knocking out the NPY gene in XY mice [98101] and comparing the knockout (KO) to WT XY or adding a transgenic copy of the candidate NPY gene in mice with one X chromosome (e.g., XO or XY*X) and comparing to controls without the transgene [73, 102]. It would be best to start with global KO or transgenic, where the genetic manipulation affects all tissues, but tissue-specific gene targeting or transgenesis would ultimately be a powerful demonstration of the effect of the NPY in the target tissue. Transgenic lines encoding autosomal copes of NPY genes (Uty, Kdm5d, Ddx3y, Usp9y, Ubely, and Eif2s3y) were developed in the Burgoyne Lab and are being backcrossed to B6 in the Arnold Lab.

It is possible but less likely that multi-copy NPY genes account for the SCE. Manipulating these genes is more problematic. One option is to knock down the expression using a transgene that delivers small interfering RNAs such as has been successful for the multi-copy NPY gene Sly [103].

Investigators who have obtained evidence for NPY involvement might consider using crosses in Additional file 4 to map the NPY genes that underlie the direct SCE to specific regions of the Y. The first cross involves a commercially available Sxr a mutation that involves translocation of Y short arm (Yp) genes to the tip of the X or Y PAR and will establish if the SCE involves Yp genes that map to Sxr a [104]. If this is not the case, the second cross utilizing a Y deletion (Yd1) should provide confirmation that multi-copy NPY genes are involved. A contact e-mail is provided in Additional file 4 for those wishing to use this Yd1 deletion.

Interpreting cases of dual NPX and NPY effects obtained with Cross A or B

Here, we consider the strategies for identifying the genes underlying a direct SCE when NPX and NPY have similar effects in Cross A and/or B. The prime candidate genes are the ancestral X-Y gene pairs Uba1y/Uba1, Kdm5d/c, Zfy/x, and Uty/x (Utx A.K.A. Kdm6a), Usp9y/x, Ddx3y/x, and Eif2s3y/x; Table 6). As discussed in the section above, “Candidate genes for direct SCEs”, the X partners of these gene pairs are widely expressed dosage-sensitive regulatory genes [19, 20]. The conservation on the Y chromosome of the Y partner gene during evolution is thought to have been driven by dosage balance of expression and function of the X and Y gametologs, which implies equivalent function of the two genes. However, over time, there were varying degrees of sequence divergence between the X and Y copies, with the Y copies often acquiring some specific functions. Thus, a central unresolved question is whether the X and Y partner genes have similar or divergent function in different tissues and life stages. Limited evidence to date indicates that the function of the X and Y genes outside of the testes is different in some cases such as Uty/Utx [105].

The RNAseq data provides a list of the X and Y genes that are expressed in the target tissue, so it is reasonable to prioritize any X-Y gene pairs. If any X genes are known to show linkage to the trait, they would have a high priority whether or not they are a member of an X-Y gene pair. For X-Y gene pairs, the relative abundance of X + X transcripts in XX females compared to X + Y transcript abundance in males are not very informative when the X and Y transcripts have diverged such that the encoded proteins are not equivalent in activity and/or function. Once again, the most direct approaches to identifying the X and Y genes involved are gene targeting and transgenesis.

Nevertheless, the possible balance of effects of X-Y pairs could lead to an unusual scenario, in which Crosses A and B show effects of NPX dose, or NPY dose, when in fact, the study of FCG mice shows no direct SCE. In this case, a likely scenario is that both members of an X-Y gene pair have effects on the phenotype but that the effects are balanced, and therefore produce no phenotypic difference in XX vs. XY. Similarly, when gene dose is manipulated through knockout of single copies of X-Y gene pairs, reducing the dose of both X and Y partners could show a phenotypic effect that is balanced and does not contribute to sex differences in phenotype. The balance of X and Y partner genes could be dynamic, however, changing under different life conditions (stress, disease, developmental stage, tissue type, and hormone levels) [50].

Looking for direct SCEs when there is no sex difference

Most investigators will be drawn to the study of FCG mice if they hypothesize a direct SCE because classical experiments of hormone removal and replacement fail to explain a known sex difference in phenotype [69]. However, FCG mice can be informative even if a sex difference in the phenotype is not established. In some cases, two sex-biasing factors typical of one sex (e.g., female levels of estradiol and a second X chromosome [47]) can reduce or eliminate the effects of the other [46, 106]. Reducing the effects of one sex-biasing factor can therefore “unmask” the effects of another. The “compensatory” actions of hormones and direct SCEs provides a rationale for comparing groups of FCG mice even if there is no overt sex difference in a phenotype [106]. In published studies, FCG mice have often been gonadectomized to allow comparison of groups under conditions in which the levels of gonadal hormones are equal, to reduce the possibility of gonadal hormonal confounds for direct SCEs. Direct SCEs discovered in gonadectomized (GDX) mice are sometimes, however, reduced if the mice have gonads or are treated with hormones after GDX [107]. Direct SCEs that are smaller in the presence of gonads or gonadal hormones are potentially important because various life events (aging, stress, disease) can result in reduced levels of gonadal hormones, which can result in the emergence of direct SCEs that contribute to sex differences in physiology. The interaction of direct SCEs and hormones, when the effect of one sex-biased variable is conditioned by the level of the other factor, indicates that the two types of variables (sex chromosome genes, gonadal hormones) have convergent actions on gene pathways influencing emergent phenotypes including the disease [45]. The molecular nature of this interaction is unstudied to date.

Conclusions

Our attempt here has been to provide the basis for a logical dissection of sex chromosome effects on any phenotype in mouse. The decision tree that summarizes this approach is shown in Fig. 4. Because the difference in the XX and XY genomes is fundamentally a difference in copy number of large segments of X and Y DNA, it makes sense to start by varying the number of these segments in combination or in isolation, to narrow down the segments that make XX and XY mice different. A progressive narrowing of choices can ultimately lead to the discovery of new genetic factors that make males and females different. The approaches suggested here have the advantage that they mimic the natural differences between XX and XY tissues. Other methods to search for X chromosome effects, for example, linkage mapping for specific traits, are not equivalent and may not detect differences caused by one vs. two X chromosomes. For example, study populations may not have any variation in the X genes that contribute to direct SCEs or variation in gene sequence may not mimic effects of X chromosome number. Similarly, studying the effects of different strain origin of the Y chromosome (e.g., by comparing consomic Y strains) may uncover effects of Y chromosome variation but may not mimic the effects of the presence vs. absence of the Y chromosome as it relates to sex differences in phenotype.

Fig. 4
figure 4

Logic tree for genetic dissection of direct sex chromosome effects

The approaches suggested here have already demonstrated that both X and Y segments cause differences in phenotype [11, 50]. Specific Y genes have been identified that control specific aspects of spermatogenesis [73, 102, 104, 108, 109], but to date, no specific X gene has been identified that causes a sex chromosome effect. Because of the advent of more efficient gene targeting methods, we expect this situation to change soon. It will be interesting to discover which X imprinted or escapee genes cause differences in physiology and disease, and how these and Y genes interact with gonadal hormones to cause emergent sex differences in phenotype.

Abbreviations

ANOVA:

Analysis of variance

CEN:

Centromere

F:

Female

FCG:

Four core genotypes mouse model

KO:

Knockout

M:

Male

NPX:

Non-PAR region of the X chromosome

NPX+:

Minute region of the NPX near the PAR

NPY:

Non-PAR region of the Y chromosome

Paf :

Patchy fur mutation

PAR:

Pseudoautosomal region

RNAseq:

RNA sequencing

SCE:

Sex chromosome effect

WT:

Wild-type

Xi :

Inactive X chromosome

Xm :

X chromosome inherited from the mother

Xp :

X chromosome inherited from the father

XXF:

XX mice with female gonadal sex (developed ovaries)

XXM:

XX mice with male gonadal sex (developed testes)

XYF:

XY mice with female gonadal sex (developed ovaries)

XYM:

XY mice with male gonadal sex (developed testes)

Y*:

Specific variant of the Y chromosome with a normal NPY but abnormal PAR

Yˉ:

Y chromosome deleted for Sry

References

  1. Capel B, Lovell-Badge R. The Sry gene and sex determination in mammals. Adv Dev Biol. 1993;2:1–35.

    Google Scholar 

  2. Gubbay J, Collignon J, Koopman P, Capel B, Economou A, Munsterberg A, Vivian N, Goodfellow P, Lovell-Badge R. A gene mapping to the sex-determining region of the mouse Y chromosome is a member of a novel family of embryonically expressed genes. Nature. 1990;346:245–50.

    Article  CAS  PubMed  Google Scholar 

  3. Koopman P, Gubbay J, Vivian N, Goodfellow P, Lovell-Badge R. Male development of chromosomally female mice transgenic for Sry. Nature. 1991;351:117–21.

    Article  CAS  PubMed  Google Scholar 

  4. Sinclair AH, Berta P, Palmer MS, Hawkins JR, Griffiths BL, Smith MJ, Foster JW, Frischauf A-M, Lovell-Badge R, Goodfellow PN. A gene from the human sex-determining region encoding a protein with homology to a conserved DNA binding motif. Nature. 1990;346:240–4.

    Article  CAS  PubMed  Google Scholar 

  5. Wilhelm D, Palmer S, Koopman P. Sex determination and gonadal development in mammals. Physiol Rev. 2007;87:1–28.

    Article  CAS  PubMed  Google Scholar 

  6. Arnold AP. Sex chromosomes and brain gender. Nat Rev Neurosci. 2004;5:701–8.

    Article  CAS  PubMed  Google Scholar 

  7. Burgoyne PS. A Y-chromosomal effect on blastocyst cell number in mice. Development. 1993;117:341–5.

    CAS  PubMed  Google Scholar 

  8. Burgoyne PS, Thornhill AR, Kalmus Boudreau S, Darling SM, Bishop CE, Evans EP. The genetic basis of XX-XY differences present before sex differentiation in the mouse. Philosophical Trans Royal Soc London (Biology). 1995;350:253–61.

    Article  CAS  Google Scholar 

  9. Corre C, Friedel M, Vousden DA, Metcalf A, Spring S, Qiu LR, Lerch JP, Palmert MR. Separate effects of sex hormones and sex chromosomes on brain structure and function revealed by high-resolution magnetic resonance imaging and spatial navigation assessment of the four core genotype mouse model. Brain Struct Funct. 2014. doi:10.1007/s00429-014-0952-0.

    PubMed  Google Scholar 

  10. Thornhill AR, Burgoyne PS. A paternally imprinted X chromosome retards the development of the early mouse embryo. Development. 1993;118:171–4.

    CAS  PubMed  Google Scholar 

  11. Arnold AP, Reue K, Eghbali M, Vilain E, Chen X, Ghahramani N, Itoh Y, Li J, Link JC, Ngun T, Williams-Burris SM. The importance of having two X chromosomes. Philos Trans R Soc Lond B Biol Sci. 2016;371:20150113.

    Article  PubMed  Google Scholar 

  12. Arnold AP, Chen X. What does the “four core genotypes” mouse model tell us about sex differences in the brain and other tissues? Front Neuroendocrinol. 2009;30:1–9.

    Article  PubMed  Google Scholar 

  13. Cox KH, Bonthuis PJ, Rissman EF. Mouse model systems to study sex chromosome genes and behavior: relevance to humans. Front Neuroendocrinol. 2014;35:405–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Charlesworth B, Charlesworth D. The degeneration of Y chromosomes. Philos Trans R Soc Lond B Biol Sci. 2000;355:1563–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Graves JA. Sex chromosome specialization and degeneration in mammals. Cell. 2006;124:901–14.

    Article  PubMed  CAS  Google Scholar 

  16. Burgoyne PS. The mammalian Y chromosome: a new perspective. Bioessays. 1998;20:363–6.

    Article  CAS  PubMed  Google Scholar 

  17. Graves JA. The origin and function of the mammalian Y chromosome and Y-borne gene—an evolving understanding. Bioessays. 1995;17:311–20.

    Article  CAS  PubMed  Google Scholar 

  18. Lahn BT, Page DC. Four evolutionary strata on the human X chromosome. Science. 1999;286:964–7.

    Article  CAS  PubMed  Google Scholar 

  19. Bellott DW, Hughes JF, Skaletsky H, Brown LG, Pyntikova T, Cho TJ, Koutseva N, Zaghlul S, Graves T, Rock S, et al. Mammalian Y chromosomes retain widely expressed dosage-sensitive regulators. Nature. 2014;508:494–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cortez D, Marin R, Toledo-Flores D, Froidevaux L, Liechti A, Waters PD, Grutzner F, Kaessmann H. Origins and functional evolution of Y chromosomes across mammals. Nature. 2014;508:488–93.

    Article  CAS  PubMed  Google Scholar 

  21. Skaletsky H, Kuroda-Kawaguchi T, Minx PJ, Cordum HS, Hillier L, Brown LG, Repping S, Pyntikova T, Ali J, Bieri T, et al. The male-specific region of the human Y chromosome is a mosaic of discrete sequence classes. Nature. 2003;423:825–37.

    Article  CAS  PubMed  Google Scholar 

  22. Soh YQ, Alfoldi J, Pyntikova T, Brown LG, Graves T, Minx PJ, Fulton RS, Kremitzki C, Koutseva N, Mueller JL, et al. Sequencing the mouse Y chromosome reveals convergent gene acquisition and amplification on both sex chromosomes. Cell. 2014;159:800–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cocquet J, Ellis PJ, Mahadevaiah SK, Affara NA, Vaiman D, Burgoyne PS. A genetic basis for a postmeiotic x versus y chromosome intragenomic conflict in the mouse. PLoS Genet. 2012;8:e1002900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Burgoyne PS. Genetic homology and crossing over in the X and Y chromosomes of mammals. Hum Genet. 1982;61:85–90.

    Article  CAS  PubMed  Google Scholar 

  25. Burgoyne PS. Mammalian X and Y crossover. Nature. 1986;319:258–9.

    Article  CAS  PubMed  Google Scholar 

  26. Keitges EA, Rivest M, Siniscalco M, Gartler SM. X-linkage of steroid sulfatase in the mouse is evidence for a functional Y-linked allele. Nature. 1985;315:226–7.

    Article  CAS  PubMed  Google Scholar 

  27. Rouyer F, Simmler MC, Johnsson C, Vergnaud G, Cooke HJ, Weissenbach J. A gradient of sex linkage in the pseudoautosomal region of the human sex chromosomes. Nature. 1986;319:291–5.

    Article  CAS  PubMed  Google Scholar 

  28. Disteche CM. Dosage compensation of the sex chromosomes. Annu Rev Genet. 2012;46:537–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Itoh Y, Melamed E, Yang X, Kampf K, Wang S, Yehya N, Van Nas A, Replogle K, Band MR, Clayton DF, et al. Dosage compensation is less effective in birds than in mammals. J Biol. 2007;6:2.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Wu H, Luo J, Yu H, Rattner A, Mo A, Wang Y, Smallwood PM, Erlanger B, Wheelan SJ, Nathans J. Cellular resolution maps of X chromosome inactivation: implications for neural development, function, and disease. Neuron. 2014;81:103–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Migeon BR. The single active X in human cells: evolutionary tinkering personified. Hum Genet. 2011;130:281–93.

    Article  PubMed  Google Scholar 

  32. Deng X, Berletch JB, Nguyen DK, Disteche CM. X chromosome regulation: diverse patterns in development, tissues and disease. Nat Rev Genet. 2014;15:367–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Migeon BR. Females are mosaics: X inactivation and sex differences in disease. New York: Oxford University Press; 2007.

    Google Scholar 

  34. Itoh Y, Arnold AP. Are females more variable than males in gene expression? Meta-analysis of microarray datasets. Biol Sex Differ. 2015;6:18.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Lemos B, Araripe LO, Hartl DL. Polymorphic Y chromosomes harbor cryptic variation with manifold functional consequences. Science. 2008;319:91–3.

    Article  CAS  PubMed  Google Scholar 

  36. Lemos B, Branco AT, Hartl DL. Epigenetic effects of polymorphic Y chromosomes modulate chromatin components, immune response, and sexual conflict. Proc Natl Acad Sci U S A. 2010;107:15826–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wijchers PJ, Festenstein RJ. Epigenetic regulation of autosomal gene expression by sex chromosomes. Trends Genet. 2011;27:132–40.

    Article  CAS  PubMed  Google Scholar 

  38. Wijchers PJ, Yandim C, Panousopoulou E, Ahmad M, Harker N, Saveliev A, Burgoyne PS, Festenstein R. Sexual dimorphism in mammalian autosomal gene regulation is determined not only by Sry but by sex chromosome complement as well. Dev Cell. 2010;19:477–84.

    Article  CAS  PubMed  Google Scholar 

  39. Morohashi K. The ontogenesis of the steroidogenic tissues. Genes Cells. 1997;2:95–106.

    Article  CAS  PubMed  Google Scholar 

  40. Parker KL, Rice DA, Lala DS, Ikeda Y, Luo X, Wong M, Bakke M, Zhao L, Frigeri C, Hanley NA, et al. Steroidogenic factor 1: an essential mediator of endocrine development. Recent Prog Horm Res. 2002;57:19–36.

    Article  CAS  PubMed  Google Scholar 

  41. Grgurevic N, Budefeld T, Rissman EF, Tobet SA, Majdic G. Aggressive behaviors in adult SF-1 knockout mice that are not exposed to gonadal steroids during development. Behav Neurosci. 2008;122:876–84.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Majdic G, Young M, Gomez-Sanchez E, Anderson P, Szczepaniak LS, Dobbins RL, McGarry JD, Parker KL. Knockout mice lacking steroidogenic factor 1 are a novel genetic model of hypothalamic obesity. Endocrinology. 2002;143:607–14.

    Article  CAS  PubMed  Google Scholar 

  43. Budefeld T, Grgurevic N, Tobet SA, Majdic G. Sex differences in brain developing in the presence or absence of gonads. Dev Neurobiol. 2008;68:981–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. De Vries GJ, Rissman EF, Simerly RB, Yang L-Y, Scordalakes EM, Auger C, Swain A, Lovell-Badge R, Burgoyne PS, Arnold AP. A model system for study of sex chromosome effects on sexually dimorphic neural and behavioral traits. J Neurosci. 2002;22:9005–14.

    PubMed  Google Scholar 

  45. Arnold AP, Chen X, Link JC, Itoh Y, Reue K. Cell-autonomous sex determination outside of the gonad. Dev Dyn. 2013;242:371–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. De Vries GJ. Minireview: sex differences in adult and developing brains: compensation, compensation, compensation. Endocrinology. 2004;145:1063–8.

    Article  PubMed  CAS  Google Scholar 

  47. Chen X, McClusky R, Chen J, Beaven SW, Tontonoz P, Arnold AP, Reue K. The number of x chromosomes causes sex differences in adiposity in mice. PLoS Genet. 2012;8:e1002709.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Li J, Chen X, McClusky R, Ruiz-Sundstrom M, Itoh Y, Umar S, Arnold AP, Eghbali M. The number of X chromosomes influences protection from cardiac ischaemia/reperfusion injury in mice: one X is better than two. Cardiovasc Res. 2014;102:375–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Smith-Bouvier DL, Divekar AA, Sasidhar M, Du S, Tiwari-Woodruff SK, King JK, Arnold AP, Singh RR, Voskuhl RR. A role for sex chromosome complement in the female bias in autoimmune disease. J Exp Med. 2008;205:1099–108.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Chen X, McClusky R, Itoh Y, Reue K, Arnold AP. X and Y chromosome complement influence adiposity and metabolism in mice. Endocrinology. 2013;154:1092–104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Itoh Y, Mackie R, Kampf K, Domadia S, Brown JD, O’Neill R, Arnold AP. Four core genotypes mouse model: localization of the Sry transgene and bioassay for testicular hormone levels. BMC Res Notes. 2015;8:69.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Manwani B, Bentivegna K, Benashski SE, Venna VR, Xu Y, Arnold AP, McCullough LD. Sex differences in ischemic stroke sensitivity are influenced by gonadal hormones, not by sex chromosome complement. J Cereb Blood Flow Metab. 2015;35:221–9.

    Article  CAS  PubMed  Google Scholar 

  53. Du S, Itoh N, Askarinam S, Hill H, Arnold AP, Voskuhl RR. XY sex chromosome complement, compared with XX, in the CNS confers greater neurodegeneration during experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A. 2014;111:2806–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mahadevaiah SK, Lovell-Badge R, Burgoyne PS. Tdy-negative XY, XXY and XYY female mice: breeding data and synaptonemal complex analysis. J Reprod Fertil. 1993;97:151–60.

    Article  CAS  PubMed  Google Scholar 

  55. Vernet N, Szot M, Mahadevaiah SK, Ellis PJ, Decarpentrie F, Ojarikre OA, Rattigan A, Taketo T, Burgoyne PS. The expression of Y-linked Zfy2 in XY mouse oocytes leads to frequent meiosis 2 defects, a high incidence of subsequent early cleavage stage arrest and infertility. Development. 2014;141:855–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kopsida E, Lynn PM, Humby T, Wilkinson LS, Davies W. Dissociable effects of Sry and sex chromosome complement on activity, feeding and anxiety-related behaviours in mice. PLoS One. 2013;8:e73699.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Burgoyne PS. Genetics of XX and XO sex reversal in the mouse. In: Wachtel S, editor. Evolutionary mechanisms of sex determination. Boca Raton, Florida: CRC Press Inc; 1988. p. 161–9.

    Google Scholar 

  58. Hunt PA, Worthman C, Levinson H, Stallings J, LeMaire R, Mroz K, Park C, Handel MA. Germ cell loss in the XXY male mouse: altered X chromosome dosage affects prenatal development. Mol Reprod Dev. 1998;49:101–11.

    Article  CAS  PubMed  Google Scholar 

  59. Gatewood JD, Wills A, Shetty S, Xu J, Arnold AP, Burgoyne PS, Rissman EF. Sex chromosome complement and gonadal sex influence aggressive and parental behaviors in mice. J Neurosci. 2006;26:2335–42.

    Article  CAS  PubMed  Google Scholar 

  60. Holaskova I, Franko J, Goodman RL, Arnold AP, Schafer R. The XX sex chromosome complement is required in male and female mice for enhancement of immunity induced by exposure to 3,4-dichloropropionanilide. Am J Reprod Immunol. 2015;74:136–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wagner CK, Xu J, Pfau JL, Quadros PS, De Vries GJ, Arnold AP. Neonatal mice possessing an Sry transgene show a masculinized pattern of progesterone receptor expression in the brain independent of sex chromosome status. Endocrinology. 2004;145:1046–9.

    Article  CAS  PubMed  Google Scholar 

  62. McPhie-Lalmansingh AA, Tejada LD, Weaver JL, Rissman EF. Sex chromosome complement affects social interactions in mice. Horm Behav. 2008;54:565–70.

    Article  PubMed  PubMed Central  Google Scholar 

  63. McCullough LD, Mirza MA, Xu Y, Bentivegna K, Steffens EB, Ritzel R, Liu F: Stroke sensitivity in the aged: sex chromosome complement vs. gonadal hormones. Aging (Albany NY). 2016. Doi: 10.18632/aging.100997.

  64. Palaszynski KM, Smith DL, Kamrava S, Burgoyne PS, Arnold AP, Voskuhl RR. A yin-yang effect between sex chromosome complement and sex hormones on the immune response. Endocrinology. 2005;146:3280–5.

    Article  CAS  PubMed  Google Scholar 

  65. Sasidhar MV, Itoh N, Gold SM, Lawson GW, Voskuhl RR. The XX sex chromosome complement in mice is associated with increased spontaneous lupus compared with XY. Ann Rheum Dis. 2012;71:1418–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Arnold AP. The organizational-activational hypothesis as the foundation for a unified theory of sexual differentiation of all mammalian tissues. Horm Behav. 2009;55:570–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Chen X, Grisham W, Arnold AP. X chromosome number causes sex differences in gene expression in adult mouse striatum. Eur J Neurosci. 2009;29:768–76.

    Article  PubMed  Google Scholar 

  68. Gioiosa L, Chen X, Watkins R, Umeda EA, Arnold AP. Sex chromosome complement affects nociception and analgesia in newborn mice. J Pain. 2008;9:962–9.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Becker JB, Arnold AP, Berkley KJ, Blaustein JD, Eckel LA, Hampson E, Herman JP, Marts S, Sadee W, Steiner M, et al. Strategies and methods for research on sex differences in brain and behavior. Endocrinology. 2005;146:1650–73.

    Article  CAS  PubMed  Google Scholar 

  70. Eicher EM, Hale DW, Hunt PA, Lee BK, Tucker PK, King TR, Eppig JT, Washburn LL. The mouse Y* chromosome involves a complex rearrangement, including interstitial positioning of the pseudoautosomal region. Cytogenet Cell Genet. 1991;57:221–30.

    Article  CAS  PubMed  Google Scholar 

  71. Burgoyne PS, Mahadevaiah SK, Perry J, Palmer SJ, Ashworth A. The Y* rearrangement in mice: new insights into a perplexing PAR. Cytogenet Cell Genet. 1998;80:37–40.

    Article  CAS  PubMed  Google Scholar 

  72. Wolstenholme JT, Rissman EF, Bekiranov S. Sexual differentiation in the developing mouse brain: contributions of sex chromosome genes. Genes Brain Behav. 2013;12:166–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Vernet N, Mahadevaiah SK, Yamauchi Y, Decarpentrie F, Mitchell MJ, Ward MA, Burgoyne PS. Mouse Y-linked Zfy1 and Zfy2 are expressed during the male-specific interphase between meiosis I and meiosis II and promote the 2nd meiotic division. PLoS Genet. 2014;10:e1004444.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Yamauchi Y, Riel JM, Ruthig V, Ward MA. Mouse Y-encoded transcription factor Zfy2 is essential for sperm formation and function in assisted fertilization. PLoS Genet. 2015;11:e1005476.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Hunt PA. Survival of XO mouse fetuses: effect of parental origin of the X chromosome or uterine environment? Development. 1991;111:1137–41.

    CAS  PubMed  Google Scholar 

  76. Lue YH, Wang C, Liu PY, Erkilla K, Swerdloff RS. Insights into the pathogenesis of XXY phenotype from comparison of the clinical syndrome with an experimental XXY mouse model. Pediatr Endocrinol Rev. 2010;8 Suppl 1:140–4.

    PubMed  Google Scholar 

  77. Burgoyne PS, Biggers JD. The consequences of X-dosage deficiency in the germ line: impaired development in vitro of preimplantation embryos from XO mice. Dev Biol. 1976;51:109–17.

    Article  CAS  PubMed  Google Scholar 

  78. Burgoyne PS, Tam PPL, Evans EP. Retarded development of XO conceptuses during early pregnancy in the mouse. J Reprod Fertil. 1983;68:387–93.

    Article  CAS  PubMed  Google Scholar 

  79. Jamieson RV, Tan S-S, Tam PPL. Retarded postimplantation development of X0 mouse embryos: impact of the parental origin of the monosomic X chromosome. Dev Biol. 1998;201:13–25.

    Article  CAS  PubMed  Google Scholar 

  80. Bonthuis PJ, Huang WC, Stacher Horndli CN, Ferris E, Cheng T, Gregg C. Noncanonical genomic imprinting effects in offspring. Cell Rep. 2015;12:979–91.

    Article  CAS  PubMed  Google Scholar 

  81. Lee JT, Bartolomei MS. X-inactivation, imprinting, and long noncoding RNAs in health and disease. Cell. 2013;152:1308–23.

    Article  CAS  PubMed  Google Scholar 

  82. Mak W, Nesterova TB, de Napoles M, Appanah R, Yamanaka S, Otte AP, Brockdorff N. Reactivation of the paternal X chromosome in early mouse embryos. Science. 2004;303:666–9.

    Article  CAS  PubMed  Google Scholar 

  83. Okamoto I, Otte AP, Allis CD, Reinberg D, Heard E. Epigenetic dynamics of imprinted X inactivation during early mouse development. Science. 2004;303:644–9.

    Article  CAS  PubMed  Google Scholar 

  84. Banzai M, Omoe K, Ishikawa H, Endo A. Viability, development and incidence of chromosome anomalies of preimplantation embryos from XO mice. Cytogen Cell Genet. 1995;70:273–7.

    Article  CAS  Google Scholar 

  85. Davies W, Isles A, Smith R, Karunadasa D, Burrmann D, Humby T, Ojarikre O, Biggin C, Skuse D, Burgoyne P, Wilkinson L. Xlr3b is a new imprinted candidate for X-linked parent-of-origin effects on cognitive function in mice. Nat Genet. 2005;37:625–9.

    Article  CAS  PubMed  Google Scholar 

  86. Raefski AS, O’Neill MJ. Identification of a cluster of X-linked imprinted genes in mice. Nat Genet. 2005;37:620–4.

    Article  CAS  PubMed  Google Scholar 

  87. Babak T, DeVeale B, Tsang EK, Zhou Y, Li X, Smith KS, Kukurba KR, Zhang R, Li JB, van der Kooy D, et al. Genetic conflict reflected in tissue-specific maps of genomic imprinting in human and mouse. Nat Genet. 2015;47:544–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Hook EB, Warburton D. Turner syndrome revisited: review of new data supports the hypothesis that all viable 45, X cases are cryptic mosaics with a rescue cell line, implying an origin by mitotic loss. Hum Genet. 2014;133:417–24.

    Article  CAS  PubMed  Google Scholar 

  89. Berletch JB, Ma W, Yang F, Shendure J, Noble WS, Disteche CM, Deng X. Escape from X inactivation varies in mouse tissues. PLoS Genet. 2015;11:e1005079.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Ellis PJ, Bacon J, Affara NA. Association of Sly with sex-linked gene amplification during mouse evolution: a side effect of genomic conflict in spermatids? Hum Mol Genet. 2011;20:3010–21.

    Article  CAS  PubMed  Google Scholar 

  91. Yang F, Babak T, Shendure J, Disteche CM. Global survey of escape from X inactivation by RNA-sequencing in mouse. Genome Res. 2010;20:614–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Bonthuis PJ, Rissman EF. Neural growth hormone implicated in body weight sex differences. Endocrinology. 2013;154:3826–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Lopes AM, Burgoyne PS, Ojarikre A, Bauer J, Sargent CA, Amorim A, Affara NA. Transcriptional changes in response to X chromosome dosage in the mouse: implications for X inactivation and the molecular basis of Turner Syndrome. BMC Genomics. 2010;11:82.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Xu J, Deng X, Disteche CM. Sex-specific expression of the X-linked histone demethylase gene Jarid1c in brain. PLoS One. 2008;3:e2553.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Xu J, Deng X, Watkins R, Disteche CM. Sex-specific differences in expression of histone demethylases Utx and Uty in mouse brain and neurons. J Neurosci. 2008;28:4521–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Xu J, Taya S, Kaibuchi K, Arnold AP. Sexually dimorphic expression of Usp9x is related to sex chromosome complement in adult mouse brain. Eur J Neurosci. 2005;21:3017–22.

    Article  CAS  PubMed  Google Scholar 

  97. Xu J, Watkins R, Arnold AP. Sexually dimorphic expression of the X-linked gene Eif2s3x mRNA but not protein in mouse brain. Gene Expr Patterns. 2006;6:146–55.

    Article  CAS  PubMed  Google Scholar 

  98. Wang H, Hu YC, Markoulaki S, Welstead GG, Cheng AW, Shivalila CS, Pyntikova T, Dadon DB, Voytas DF, Bogdanove AJ, et al. TALEN-mediated editing of the mouse Y chromosome. Nat Biotechnol. 2013;31:530–2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Wang H, Yang H, Shivalila CS, Dawlaty MM, Cheng AW, Zhang F, Jaenisch R. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell. 2013;153:910–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Matsubara Y, Kato T, Kashimada K, Tanaka H, Zhi Z, Ichinose S, Mizutani S, Morio T, Chiba T, Ito Y, et al. TALEN-mediated gene disruption on Y chromosome reveals critical role of EIF2S3Y in mouse spermatogenesis. Stem Cells Dev. 2015;24:1164–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Singh P, Schimenti JC, Bolcun-Filas E. A mouse geneticist’s practical guide to CRISPR applications. Genetics. 2015;199:1–15.

    Article  CAS  PubMed  Google Scholar 

  102. Mazeyrat S, Saut N, Grigoriev V, Mahadevaiah SK, Ojarikre OA, Rattigan A, Bishop C, Eicher EM, Mitchell MJ, Burgoyne PS. A Y-encoded subunit of the translation initiation factor Eif2 is essential for mouse spermatogenesis. Nat Genet. 2001;29:49–53.

    Article  CAS  PubMed  Google Scholar 

  103. Cocquet J, Ellis PJ, Yamauchi Y, Mahadevaiah SK, Affara NA, Ward MA, Burgoyne PS. The multicopy gene Sly represses the sex chromosomes in the male mouse germline after meiosis. PLoS Biol. 2009;7:e1000244.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Vernet N, Mahadevaiah SK, Decarpentrie F, Longepied G, de Rooij DG, Burgoyne PS, Mitchell MJ. Mouse Y-encoded transcription factor Zfy2 is essential for sperm head remodelling and sperm tail development. PLoS One. 2016;11:e0145398.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Shpargel KB, Sengoku T, Yokoyama S, Magnuson T. UTX and UTY demonstrate histone demethylase-independent function in mouse embryonic development. PLoS Genet. 2012;8:e1002964.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Arnold AP. Conceptual frameworks and mouse models for studying sex differences in physiology and disease: why compensation changes the game. Exp Neurol. 2014;259:2–9.

    Article  PubMed  Google Scholar 

  107. Chen X, Wang L, Loh DH, Colwell CS, Tache Y, Reue K, Arnold AP. Sex differences in diurnal rhythms of food intake in mice caused by gonadal hormones and complement of sex chromosomes. Horm Behav. 2015;75:55–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Royo H, Polikiewicz G, Mahadevaiah SK, Prosser H, Mitchell M, Bradley A, de Rooij DG, Burgoyne PS, Turner JM. Evidence that meiotic sex chromosome inactivation is essential for male fertility. Curr Biol. 2010;20:2117–23.

    Article  CAS  PubMed  Google Scholar 

  109. Vernet N, Mahadevaiah SK, Ojarikre OA, Longepied G, Prosser HM, Bradley A, Mitchell MJ, Burgoyne PS. The Y-encoded gene Zfy2 acts to remove cells with unpaired chromosomes at the first meiotic metaphase in male mice. Curr Biol. 2011;21:787–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Wright WE, Sassoon DA, Lin VK. Myogenin, a factor regulating myogenesis, has a domain homologous to MyoD. Cell. 1989;56:607–17.

    Article  CAS  PubMed  Google Scholar 

  111. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2001;25:402–8.

    Article  CAS  PubMed  Google Scholar 

  112. Burgoyne PS, Evans EP. A high frequency of XO offspring from X(Paf)Y* male mice: evidence that the Paf mutation involves an inversion spanning the X PAR boundary. Cytogenet Cell Genet. 2000;91:57–61.

    Article  CAS  PubMed  Google Scholar 

  113. Lane PW, Davisson MT. Patchy fur (Paf), a semidominant X-linked gene associated with a high level of X-Y nondisjunction in male mice. J Hered. 1990;81:43–50.

    CAS  PubMed  Google Scholar 

  114. Koehler KE, Millie EA, Cherry JP, Burgoyne PS, Evans EP, Hunt PA, Hassold TJ. Sex-specific differences in meiotic chromosome segregation revealed by dicentric bridge resolution in mice. Genetics. 2002;162:1367–79.

    PubMed  PubMed Central  Google Scholar 

  115. Ishikawa H, Rattigan A, Fundele R, Burgoyne PS. Effects of sex chromosome dosage on placental size in mice. Biol Reprod. 2003;69:483–8.

    Article  CAS  PubMed  Google Scholar 

  116. Mahadevaiah SK, Odorisio T, Elliott DJ, Rattigan A, Szot M, Laval SH, Washburn LL, McCarrey JR, Cattanach BM, Lovell-Badge R, Burgoyne PS. Mouse homologues of the human AZF candidate gene RBM are expressed in spermatogonia and spermatids, and map to a Y chromosome deletion interval associated with a high incidence of sperm abnormalities. Hum Mol Genet. 1998;7:715–27.

    Article  CAS  PubMed  Google Scholar 

  117. Capel B, Rasberry C, Dyson J, Bishop CE, Simpson E, Vivian N, Lovell-Badge R, Rastan S, Cattanach BM. Deletion of Y chromosome sequences located outside the testis determining region can cause XY female sex reversal. Nat Genet. 1993;5:301–7.

    Article  CAS  PubMed  Google Scholar 

  118. Laval SH, Reed V, Blair HJ, Boyd Y. The structure of DXF34, a human X-linked sequence family with homology to a transcribed mouse Y-linked repeat. Mamm Genome. 1997;8:689–91.

    Article  CAS  PubMed  Google Scholar 

  119. Rodriguez TA, Burgoyne PS. Spermatogenic failure in male mice with four sex chromosomes. Chromosoma. 2001;110:124–9.

    Article  CAS  PubMed  Google Scholar 

  120. Touré A, Grigoriev V, Mahadevaiah SK, Rattigan A, Ojarikre OA, Burgoyne PS. A protein encoded by a member of the multicopy Ssty gene family located on the long arm of the mouse Y chromosome is expressed during sperm development. Genomics. 2004;83:140–7.

    Article  PubMed  CAS  Google Scholar 

  121. Chen X, Watkins R, Delot E, Reliene R, Schiestl RH, Burgoyne PS, Arnold AP. Sex difference in neural tube defects in p53-null mice is caused by differences in the complement of X not Y genes. Dev Neurobiol. 2008;68:265–73.

    Article  CAS  PubMed  Google Scholar 

  122. Palmer S, Perry J, Kipling D, Ashworth A. A gene spans the pseudoautosomal boundary in mice. Proc Natl Acad Sci U S A. 1997;94:12030–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Kipling D, Salido EC, Shapiro LJ, Cooke HJ. High frequency de novo alterations in the long-range genomic structure of the mouse pseudoautosomal region. Nat Genet. 1996;13:78–82.

    Article  CAS  PubMed  Google Scholar 

  124. Salido EC, Li XM, Yen PH, Martin N, Mohandas TK, Shapiro L. Cloning and expression of the mouse pseudoautosomal steroid sulphatase gene (Sts). Nat Genet. 1996;13:83–6.

    Article  CAS  PubMed  Google Scholar 

  125. Trent S, Fry JP, Ojarikre OA, Davies W. Altered brain gene expression but not steroid biochemistry in a genetic mouse model of neurodevelopmental disorder. Mol Autism. 2014;5:21.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  126. Trent S, Dean R, Veit B, Cassano T, Bedse G, Ojarikre OA, Humby T, Davies W. Biological mechanisms associated with increased perseveration and hyperactivity in a genetic mouse model of neurodevelopmental disorder. Psychoneuroendocrinology. 2013;38:1370–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Hughes JF, Skaletsky H, Koutseva N, Pyntikova T, Page DC. Sex chromosome-to-autosome transposition events counter Y-chromosome gene loss in mammals. Genome Biol. 2015;16:104.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Elliott DJ, Ma K, Kerr SM, Thakrar R, Speed R, Chandley AC, Cooke H. An RBM homologue maps to the mouse Y chromosome and is expressed in germ cells. Hum Mol Genet. 1996;5:869–74.

    Article  CAS  PubMed  Google Scholar 

  129. Dreumont N, Bourgeois CF, Lejeune F, Liu Y, Ehrmann IE, Elliott DJ, Stevenin J. Human RBMY regulates germline-specific splicing events by modulating the function of the serine/arginine-rich proteins 9G8 and Tra2-{beta}. J Cell Sci. 2010;123:40–50.

    Article  CAS  PubMed  Google Scholar 

  130. Kay GF, Ashworth A, Penny GD, Dunlop M, Swift S, Brockdorff N, Rastan S. A candidate spermatogenesis gene on the mouse Y chromosome is homologous to ubiquitin-activating enzyme E1. Nature. 1991;354:486–9.

    Article  CAS  PubMed  Google Scholar 

  131. Mitchell MJ, Woods DR, Tucker PK, Opp JS, Bishop CE. Homology of a candidate spermatogenic gene from the mouse Y chromosome to the ubiquitin-activating enzyme E1. Nature. 1991;354:483–6.

    Article  CAS  PubMed  Google Scholar 

  132. Akimoto C, Kitagawa H, Matsumoto T, Kato S. Spermatogenesis-specific association of SMCY and MSH5. Genes Cells. 2008;13:623–33.

    Article  CAS  PubMed  Google Scholar 

  133. Agulnik AI, Mitchell MJ, Lerner JL, Woods DR, Bishop CE. A mouse Y chromosome gene encoded by a region essential for spermatogenesis and expression of male-specific minor histocompatibility antigens. Hum Mol Gen. 1994;3:873–8.

    Article  CAS  PubMed  Google Scholar 

  134. Decarpentrie F, Vernet N, Mahadevaiah SK, Longepied G, Streichemberger E, Aknin-Seifer I, Ojarikre OA, Burgoyne PS, Metzler-Guillemain C, Mitchell MJ. Human and mouse ZFY genes produce a conserved testis-specific transcript encoding a zinc finger protein with a short acidic domain and modified transactivation potential. Hum Mol Genet. 2012;21:2631–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Greenfield A, Scott D, Pennisi D, Ehrmann I, Ellis P, Cooper L, Simpson E, Koopman P. An H-YDb epitope is encoded by a novel mouse Y chromosome gene. Nat Genet. 1996;14:474–8.

    Article  CAS  PubMed  Google Scholar 

  136. Brown GM, Furlong RA, Sargent CA, Erickson RP, Longepied G, Mitchell M, Jones MH, Hargreave TB, Cooke HJ, Affara NA. Characterisation of the coding sequence and fine mapping of the human DFFRY gene and comparative expression analysis and mapping to the Sxrb interval of the mouse Y chromosome of the Dffry gene. Hum Mol Gen. 1998;7:97–108.

    Article  CAS  PubMed  Google Scholar 

  137. Sun C, Skaletsky H, Birren B, Devon K, Tang Z, Silber S, Oates R, Page D. An Azoospermic man with a de novo point mutation in the Y-chromosomal gene USP9Y. Nat Genet. 1999;23:429–32.

    Article  CAS  PubMed  Google Scholar 

  138. Mazeyrat S, Saut N, Sargent CA, Grimmond S, Longepied G, Ehrmann IE, Ellis PS, Greenfield A, Affara NA, Mitchell MJ. The mouse Y chromosome interval necessary for spermatogonial proliferation is gene dense with syntenic homology to the human AZFa region. Hum Mol Genet. 1998;7:1713–24.

    Article  CAS  PubMed  Google Scholar 

  139. Ehrmann IE, Ellis PS, Mazeyrat S, Duthie S, Brockdorff N, Mattei MG, Gavin MA, Affara NA, Brown GM, Simpson E, et al. Characterization of genes encoding translation initiation factor eIF-2gamma in mouse and human: sex chromosome localisation, escape from X-inactivation and evolution. Hum Mol Gen. 1998;7:1725–37.

    Article  CAS  PubMed  Google Scholar 

  140. Xu J, Burgoyne PS, Arnold AP. Sex differences in sex chromosome gene expression in mouse brain. Hum Mol Genet. 2002;11:1409–19.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank William Davies and Rhonda Voskuhl for allowing us to use unpublished information from their labs and Yuichiro Itoh and Fanny Decarpentrie for use of figures that they generated for Additional file 2. The ideas discussed in this proposal have emerged from countless discussions with our collaborators over the last several decades, whom we acknowledge and thank.

Funding

Funding was received from the UK Medical Research Council grant U117532009 (PSB) and NIH grants (APA et al.) MH59268, DC000217, NS045966, NS043196, DK083561, HD076125, HL119886, HL131182.

Availability of data and materials

Data sharing is not applicable to this article because no datasets were generated or analyzed during the current study.

Authors’ contributions

PSB and APA wrote the manuscript. Both authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Ethics approval and consent to participate

The manuscript contains no new experimental research and therefore is not subject to approval for use of animals.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Arthur P. Arnold.

Additional files

Additional file 1:

Obtaining parents for B6 crosses used in the “Linking direct SCEs to a specific component(s) of the XX or XY complements” section. (See Additional file 2 for genotyping protocols). (DOCX 15 kb)

Additional file 2:

Progeny genotyping protocols relating to Additional file 1 [51, 7274, 110, 111]. (DOCX 25 kb)

Additional file 3:

Previously utilized MF1 crosses relevant to this review [10, 38, 67, 70, 71, 75, 7779, 85, 112115]. (DOCX 31 kb)

Additional file 4:

Crosses with Y chromosome translocations or deletions. (See chromosome diagrams in [55, 104, 116, 117]). (DOCX 20 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Burgoyne, P.S., Arnold, A.P. A primer on the use of mouse models for identifying direct sex chromosome effects that cause sex differences in non-gonadal tissues. Biol Sex Differ 7, 68 (2016). https://doi.org/10.1186/s13293-016-0115-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13293-016-0115-5

Keywords