Male-female differences in many developmental mechanisms lead to the formation of two morphologically and physiologically distinct sexes. Although this is expected for traits with prominent differences between the sexes, such as the gonads, sex-specific processes also contribute to traits without obvious male-female differences, such as the intestine. Here, we review sex differences in developmental mechanisms that operate at several levels of biological complexity – molecular, cellular, organ and organismal – and discuss how these differences influence organ formation, function and whole-body physiology. Together, the examples we highlight show that one simple way to gain a more accurate and comprehensive understanding of animal development is to include both sexes.

Male-female differences in many developmental processes lead to the formation of two morphologically and physiologically distinct sexes (Arnold, 2017, 2019). The prominent nature of some morphological differences between males and females has motivated studies aimed at uncovering the developmental mechanisms underlying the formation of these highly dimorphic traits. For example, an elegant body of work across many species has provided in-depth knowledge of the distinct genetic and hormonal mechanisms that contribute to the development of sex-specific structures such as the gonads (Arnold, 2017; Capel, 2000; Hubbard and Greenstein, 2000; Matson and Zarkower, 2012; Nef and Parada, 2000; Sekido and Lovell-Badge, 2013; Swain and Lovell-Badge, 1999; Whitworth et al., 2012; Williams and Carroll, 2009; Zarkower, 2001, 2013). Studies on sex-limited traits such as pigmentation, weapons and ornaments have similarly identified mechanisms that contribute to trait development in only one sex. For example, horns are observed only in male adult Onthophagus taurus beetles due to female-specific cell death during development (Kijimoto et al., 2010). This mechanism also explains the development of functional wings in adult male, but not female, winter moths (Nyssiodes lefuarius) (Niitsu et al., 2014). In contrast, sex-specific cell proliferation explains why males from some Xiphophorous fish species have a larger sword than females (Offen et al., 2008; Powell et al., 2021; Schartl et al., 2021). Sex-specific developmental mechanisms therefore contribute to traits with conspicuous differences between males and females.

In addition to highly dimorphic traits, it has become increasingly clear that sex differences exist in many cell types, tissues and organs that are shared between the sexes. For example, there are sex differences in neuron number, dendritic projections and size of specific brain regions in many animals (Jazin and Cahill, 2010). Although the physiological significance of these differences has not been fully elucidated, sex differences in specific neuronal populations contribute to male-female differences in diverse phenotypes such as body size, physiology and behavior (Jazin and Cahill, 2010). There are also differences between males and females across species, including humans, in the number of pancreatic insulin-producing β-cells (Marchese et al., 2015; Parchami and Kusha, 2015; van der Kroon et al., 2017), in whole-body fat storage and distribution (Karastergiou et al., 2012), in nephron number (Ecelbarger, 2016) and in conducting airways (Martin et al., 1987). Because β-cell mass is greater in females, which show a smaller average pancreas volume and duct size than males (Wang et al., 2021), this difference cannot be fully explained by the sex difference in organ size. Similarly, fat storage is ∼10% higher in female mammals despite a lower average body weight (Karastergiou et al., 2012), and the sex difference in conducting airways remains even after correcting for male-female differences in lung size (Martin et al., 1987). Thus, for many cell types, tissues and organs, sex differences cannot be fully explained by a model in which one sex has more of a specific cell type because the corresponding organ is larger. Despite sex differences in these and additional cell types and tissues (Arnold, 2019), knowledge of the underlying developmental mechanisms that establish sexual dimorphism in shared cell types, tissues and organs remains incomplete.

One reason for this knowledge gap is the frequent use of mixed- and single-sex animal groups when studying shared traits. When development is studied in a single sex, there is a significant risk that the contribution of a gene or cellular process to organ formation will be overlooked if that gene or process does not operate in the sex under investigation. Single-sex studies also carry the risk of inaccurately estimating the capacity of an organ for repair following a developmental insult, or the ability of an animal to withstand environmental fluctuations. Mixed-sex studies carry the risk of disregarding the contributions of important genes and cellular processes to the development and function of an organ because a sex-limited effect did not achieve statistical significance. Studying both sexes, and including sex as a variable in the analysis, therefore provides two simple ways for researchers to obtain a more accurate, mechanistic and detailed understanding of development. The importance of this task is shown by mounting evidence that males and females differ in many aspects of development, and in the incidence and progression of diseases related to the dysregulation of fundamental developmental processes (e.g. cell proliferation, cell death, organ repair and growth restriction) (Arnold, 2017, 2019; Clocchiatti et al., 2016; Gannon et al., 2018; Morrow, 2015; Ober et al., 2008).

One illustration of why both sexes must be included when studying development comes from high-throughput bulk and single-cell sequencing studies across multiple tissues. Gene expression differences have been documented between the sexes in many animals across tissue types (Mank, 2017; Mayne et al., 2016; Melé et al., 2015). Indeed, up to 40% of genes are differentially expressed between males and females in somatic tissues (Oliva et al., 2020). Interestingly, the degree of gene expression differences between the sexes amplifies over development, and is most pronounced in adults (Emlen et al., 2012; Mank et al., 2010; Perry et al., 2014). This suggests that gene regulatory differences between the sexes in larval, juvenile or other immature stages may be the most crucial, at least in some cases, to manifest sex differences in adults. Because development ultimately relies upon differential gene regulation, this profound male-female difference in gene expression has major implications for studies aimed at revealing genes that influence development (Lopes-Ramos et al., 2020). Given that transcriptional responses to genetic and environmental manipulations also differ between the sexes (Camus et al., 2019; Graze et al., 2018; Honjoh et al., 2017; Mitchell et al., 2016), it is clear that studying one sex does not adequately capture, or allow the prediction of, basal and induced gene expression responses in both sexes.

The overall goal of this Review is to communicate the importance of including both sexes in studies of the fundamental mechanisms underlying development. To achieve this goal, we will use specific examples from studies in a range of model and non-model animals to make general points about the benefits associated with studying both sexes. First, we discuss how sex differences at the genetic and molecular level influence developmental outcomes. We next describe how male-female differences in fundamental cellular processes shape tissue and organ properties. Third, we examine how sex differences in stem cell behavior impact organ size and capacity for repair. Finally, we highlight how sex differences in cell signaling pathways influence development at the organismal level. When taken together, the convergent patterns observed repeatedly across many animals demonstrate that lessons learned from studies in a mixed- or single-sex animal group cannot always be broadly applied to both sexes. A more accurate and mechanistic understanding of development will therefore emerge from studies that include both sexes.

A mechanistic understanding of development requires the identification of genes and gene networks that contribute to phenotypic outcomes. Emerging literature shows that the number of genes that affect a phenotype, and the relative contribution of each gene to the phenotype, often differs between the sexes. This suggests that the genetic architecture, which refers to the number and effect sizes of genetic variants that influence a given phenotype, differs between males and females for many traits (Karp et al., 2017). Although this might be expected for highly dimorphic traits (e.g. gonads, pigmentation), sex differences in genetic architecture exist even in traits without obvious male-female differences (see Table 1) (Van der Bijl and Mank, 2021).

Table 1.

International Mouse Phenotyping Consortium: traits with significant measures of sexual dimorphism

International Mouse Phenotyping Consortium: traits with significant measures of sexual dimorphism
International Mouse Phenotyping Consortium: traits with significant measures of sexual dimorphism

Sex-biased effects of sex chromosome-associated genes on development

Sex and sex differences in many species are determined entirely by environmental cues (Bachtrog et al., 2014), and in these species there are no genetic differences between females and males. However, in many species, sex is associated with sex chromosomes. Although the environment can play an important role in sex differences even in species with sex chromosomes (Badyaev, 2002), the sex chromosome complement of each sex means that by definition, some genes will affect only a single sex (Fig. 1). For example, in species with the XY sex determination system, Y chromosome genes only contribute to traits in males. Indeed, the control of pigmentation by Y chromosome genes in Poecilia parae perfectly illustrates this sex-specific phenotypic effect (Fig. 1).

Fig. 1.

Sex chromosome and sex hormone effects on sex-specific trait development. (A) The sex chromosomes, in species that have them, often provide the initial genetic trigger for phenotypic sex differences. This occurs through several different routes. First, the Y chromosome presents a male-specific region of the genome, and some dimorphisms are encoded on Y-linked genes. For example, there are five male morphs in the fish Poecilia parae, each of which displays distinct differences in color, behavior and physiology. These five morphs are each associated with a different Y chromosome, meaning that the Y ultimately encodes the male-specific traits as females of this species carry two X chromosomes (Sandkam et al., 2021). However, the coding content of the Y chromosome is very restricted in many species, and some species, in which sex is determined by X chromosome dose, have lost the Y chromosome entirely (Bachtrog et al., 2014). (B) The X chromosome differs in dose between the sexes, and although dose compensation systems in some systems equalize expression for most X-linked genes between males and females, the presence of two X chromosomes in females and one in males can play a role in sex differences. For example, male-specific wing pigmentation in Drosophila biarmipes is thought to be ultimately encoded by an X-linked gene, and transgene analysis in D. melanogaster indicates that the regulation of the gene is ultimately controlled by whether an individual has one X chromosome or two, and is independent of the sex-determination pathway itself (Galouzis and Prud'homme, 2021). Although further functional validation of this mechanism in D. biarmipes is needed, these results suggest there is a repressive interaction between the encoding gene (yellow, y) and the corresponding allele on the homologous X chromosome in females, a phenomenon known as transvection. Males have only one copy of the X chromosome, and so lack a homologous X to repress yellow. (C) Sex chromosome complement also initiates the sex-determination pathway, where some dimorphisms are controlled by sex-determination genes. For example, abdominal coloration in male Drosophila is controlled by bric-à-brac (bab), which is regulated by the Drosophila sex-determining locus doublesex (dsx) (Kopp et al., 2000). The female isoform of dsx is called DsxF and activates bab transcription, whereas the male dsx isoform, DsxM, represses bab. (D) The sex-determination cascade also leads to differences in sex hormone levels. Testosterone (T) treatment of females (which normally lack coloration) is sufficient to produce male coloration patterns in many fish (e.g. Poecilia reticulata; Gordon et al., 2012), birds (Ardia et al., 2010) and reptiles (Cox et al., 2005), indicating that sex hormones regulate these traits.

Fig. 1.

Sex chromosome and sex hormone effects on sex-specific trait development. (A) The sex chromosomes, in species that have them, often provide the initial genetic trigger for phenotypic sex differences. This occurs through several different routes. First, the Y chromosome presents a male-specific region of the genome, and some dimorphisms are encoded on Y-linked genes. For example, there are five male morphs in the fish Poecilia parae, each of which displays distinct differences in color, behavior and physiology. These five morphs are each associated with a different Y chromosome, meaning that the Y ultimately encodes the male-specific traits as females of this species carry two X chromosomes (Sandkam et al., 2021). However, the coding content of the Y chromosome is very restricted in many species, and some species, in which sex is determined by X chromosome dose, have lost the Y chromosome entirely (Bachtrog et al., 2014). (B) The X chromosome differs in dose between the sexes, and although dose compensation systems in some systems equalize expression for most X-linked genes between males and females, the presence of two X chromosomes in females and one in males can play a role in sex differences. For example, male-specific wing pigmentation in Drosophila biarmipes is thought to be ultimately encoded by an X-linked gene, and transgene analysis in D. melanogaster indicates that the regulation of the gene is ultimately controlled by whether an individual has one X chromosome or two, and is independent of the sex-determination pathway itself (Galouzis and Prud'homme, 2021). Although further functional validation of this mechanism in D. biarmipes is needed, these results suggest there is a repressive interaction between the encoding gene (yellow, y) and the corresponding allele on the homologous X chromosome in females, a phenomenon known as transvection. Males have only one copy of the X chromosome, and so lack a homologous X to repress yellow. (C) Sex chromosome complement also initiates the sex-determination pathway, where some dimorphisms are controlled by sex-determination genes. For example, abdominal coloration in male Drosophila is controlled by bric-à-brac (bab), which is regulated by the Drosophila sex-determining locus doublesex (dsx) (Kopp et al., 2000). The female isoform of dsx is called DsxF and activates bab transcription, whereas the male dsx isoform, DsxM, represses bab. (D) The sex-determination cascade also leads to differences in sex hormone levels. Testosterone (T) treatment of females (which normally lack coloration) is sufficient to produce male coloration patterns in many fish (e.g. Poecilia reticulata; Gordon et al., 2012), birds (Ardia et al., 2010) and reptiles (Cox et al., 2005), indicating that sex hormones regulate these traits.

In contrast, in many species, the X chromosome is present in two copies in females and one in males. Many species lack complete X chromosome dose compensation (Mank, 2013), and the difference in chromosome dose leads to higher expression on average in females for X-linked genes. Many other species have complete X chromosome dose compensation mechanisms, which equalize expression between the sexes (Arnold, 2017; Balaton and Brown, 2016; Deng et al., 2014), although many genes escape these mechanisms (Balaton and Brown, 2016). Because the X chromosome is present in each sex, it may influence development in males and females; however, predicting the developmental effects of X chromosome-associated genes is difficult. For example, in Drosophila, fat body loss of a growth-promoting gene called stunted, an X-linked gene, decreases body size in females but not in males (Millington et al., 2021a). In contrast, adult flies lacking the coding sequence for the X-linked gene Drosophila insulin-like peptide 6 show reduced adult weight in males but not females (Millington et al., 2021b). Both sexes must therefore be included when studying the phenotypic effects of X-linked mutations, as the size and direction of the effect cannot be predicted in advance. Indeed, the X chromosomes make a larger contribution to sex differences than expected based on their size or total gene content (Mank, 2009).

Genome-wide association studies reveal sex differences in genetic architecture of sex-specific and non-sex-specific traits

Autosomal genes also differentially affect development in males and females (Ober et al., 2008). Although the autosomes are shared equally between sexes, their vastly greater coding content makes a larger overall contribution to sex differences in the genetic architecture of development. This suggests that both sexes must be considered even in studies on developmental phenotypes associated with autosomal genes. One line of evidence supporting this suggestion emerges from genome-wide association studies (GWAS). GWAS are genomic scans looking for a correlation between genetic variants (e.g. single-nucleotide polymorphisms) and phenotypic traits, and represent a powerful big-data method to characterize the genetic architecture of specific phenotypes based on large sample numbers.

GWAS have identified sex differences in autosomal genetic architecture for traits with pronounced dimorphism, such as fat deposition (Shungin et al., 2015), body size and shape (Heid et al., 2010; Randall et al., 2013; Winkler et al., 2015), neurological traits (Lu and Cantor, 2012; Matoba et al., 2019) and disease (Box 1). These associations occur despite little evidence for allele frequency differences in genetic variants for autosomal loci between the sexes (Boraska et al., 2012). In phenotypes for which there is no obvious or known sex difference, evidence for sex differences in genetic architecture has also accumulated through sex-stratified GWAS (Graham et al., 2019; Khramtsova et al., 2019). In most cases, the sex difference in genetic architecture is due to variants that are statistically associated with a trait in one sex or the other (sex specific), despite being present in the genomes of both sexes, as discussed in the following section. Fewer genetic architecture differences arise from variants having opposing effects in the sexes (sex discordance) (Harper et al., 2020 preprint), suggesting that, although genetic architecture may differ, it does not often act in fundamentally different ways between the sexes.

Box 1. Sex differences in disease

Evidence that a large proportion of human diseases have sex differences in prevalence, age of onset, severity and/or response to treatment has been building for some time (Ober et al., 2008). Recent institutional calls to balance biomedical and clinical studies with regard to sex (Clayton and Collins, 2014) have led to a tidal wave of evidence for sex differences in disease and medically relevant phenotypes (Karp et al., 2017; Khramtsova et al., 2019; Morrow, 2015; Richardson et al., 2021).

Some of these differences are due to the X chromosome and the fact that males have an increased incidence of some X-linked diseases due to their single copy. Examples include X-linked color blindness (Xie et al., 2014) and muscular dystrophy (Towbin et al., 1993). However, the sex chromosomes do not explain the vast majority of sex differences observed in disease traits (Ge et al., 2017; Traglia et al., 2017), such as the incidence of psychiatric disorders (Bogetto et al., 1999; Mandy et al., 2012; Tükel et al., 2005) and cardiovascular disease (Haast et al., 2012; Lawlor et al., 2001). The sex chromosomes also do not explain the fact that the sexes show major differences in the pharmacokinetics for many drugs (Soldin and Mattison, 2009). Failure to account for sex differences in pharmacokinetics has led to some dangerous dosing guidelines, exemplified by the case of Ambien (zolpidem), a sleep aid where differences in metabolic clearance rates between the sexes left a significant percentage of women with lingering sedation after the intended 8 h dose (Greenblatt et al., 2000).

Moreover, detailed genetic case-control comparisons have revealed many differences in the genetic architecture of disease risk and heritability (Duncan et al., 2018; Ge et al., 2017; Traglia et al., 2017). These differences underscore the need to account for sex in clinical and pre-clinical research, and the need to develop sex-informed treatment plans for patients.

Large-scale phenotyping studies show sex-biased and sex-specific requirements for many genes in regulating development

Like GWAS, large-scale phenotyping studies also indicate widespread and unpredictable sex differences in genetic architecture for both sex chromosome- and autosomal-linked genes. For example, the International Mouse Phenotyping Consortium (IMPC) aims to map out the function of every protein-coding gene in the mouse genome (Dickinson et al., 2016). Male and female knockout mutants are subjected to a phenotyping pipeline that spans developmental, cellular, physiological, behavioral and morphological traits. Because the phenotypic effects of knockout mutations are measured in males and females, it is possible to determine the underlying differences in genetic architecture for specific traits (Karp et al., 2017).

Although previous studies mapped the effects of mutants to sex-specific phenotypes such as fertility and sterility (Connallon and Clark, 2011; Tweedie et al., 2009), the IMPC effort was the first large-scale agnostic phenotyping analysis of sex differences across a broad range of somatic traits. The results showed that not only was sexual dimorphism prevalent in phenotypes for which there was no obvious explanation (Karp et al., 2017), with up to 31% of somatic traits showing significant sex differences (Table 1) (van der Bijl and Mank, 2021), but even the variability of many traits differs between males and females (Zajitschek et al., 2020). Most importantly, roughly 17% of autosomal genes had sex-biased or sex-specific phenotypic effects (Karp et al., 2017). Of knockouts with different effects between the sexes, about 75% produced a phenotype in one sex or the other, 20% produced effects with opposing directions between the sexes and 5% produced effects in the same direction but of a different magnitude. Of note, these sex-biased and sex-specific effects were not limited to highly sex-specific organs such as the gonad. Rather, they include but are not limited to organs such as the gut, adipose tissue, the heart and the pancreas, organs where sexual dimorphism is far less obvious and highly understudied (Table 1) (Karp et al., 2017).

Together, these lines of evidence demonstrate the importance of including both sexes in studies aimed at revealing the genetic and molecular mechanisms underlying development. This principle is equally important when studying both sex chromosome- and autosomal-linked genes, and when studying developmental traits that may or may not show prominent differences between the sexes. One important risk of not including both males and females in studies is overlooking profound phenotypic consequences caused by loss of gene function. This occurs when the size of the effect does not achieve statistical significance in a mixed-sex population, or when the unaffected sex is studied in a single-sex population. In contrast, studying both sexes separately provides the opportunity to discover new genes associated with developmental outcomes that were missed in single- and mixed-sex studies.

Understanding development also requires knowledge of mechanisms that operate at the level of individual cells to specify tissue and organ properties. A rapidly growing body of evidence has identified sex differences in many fundamental cellular processes, and shown that these differences contribute to final tissue size and shape, and to organ size, anatomy and cell composition. This suggests that the cellular processes that govern final tissue and organ properties may not be shared between the sexes. Notably, these differences are not limited to reproductive organs.

Sex differences in fundamental cellular processes exist in tissues and organs without obvious sexual dimorphism

It is widely appreciated that traits with prominent sexual dimorphism (e.g. sex-limited weapons and ornaments, pigmentation and coloration) will be associated with sex-specific developmental mechanisms. Beyond these obviously dimorphic traits, however, a growing body of evidence shows that male-female differences in fundamental cellular processes exist in many organs and tissues that are shared between the sexes. In flies and other invertebrates, male-female differences have been reported in the size, shape and cell composition of the fat, wing, leg, central nervous system (CNS), gut and muscle (Stokes et al., 1994; Demir and Dickson, 2005; Rideout et al., 2010; Robinett et al., 2010; Rideout et al., 2015; Hudry et al., 2016; Regan et al., 2016; Ahmed et al., 2020; Wat et al., 2020). Importantly, many of these differences are functionally significant: sex-specific neural circuits in Drosophila promote male courtship behaviors (Demir and Dickson, 2005; Rideout et al., 2010; Robinett et al., 2010), whereas sex differences in the intestine influence reproductive output and lifespan responses to dietary nutrients (Hudry et al., 2016; Regan et al., 2016). In vertebrates, including humans, sex differences have been reported in the brain, heart, fat, pancreas, kidney and lungs (de Simone et al., 1995; Ecelbarger, 2016; Jazin and Cahill, 2010; Karastergiou et al., 2012; Legato and Legha, 2004; LoMauro and Aliverti, 2018; Marchese et al., 2015; Parchami and Kusha, 2015; Taylor et al., 2010; van der Kroon et al., 2017). Although these sex differences are largely descriptive in nature, and the underlying mechanisms have not been fully elucidated, male-female differences in phenotypes related to these organs have been noted. For example, in alignment with a higher number of pancreatic β-cells in women (Marchese et al., 2015; van der Kroon et al., 2017), glucose-stimulated insulin secretion is higher in women than in men in the context of equivalent insulin sensitivity (Basu et al., 2006, 2017; Horie et al., 2018). At least some differences noted in descriptive studies may therefore be relevant for male-female differences in development, physiology and behavior – a possibility that requires further investigation.

Of the tissues and organs without prominent sexual dimorphism, the CNS represents one of the best-studied examples of how sex differences in fundamental cellular processes shape organ properties (Ampatzis and Dermon, 2007; Jazin and Cahill, 2010; Portman, 2007; Sulston et al., 1983). For example, in Drosophila melanogaster, sex-specific programmed cell death occurs in many regions of the developing CNS (Garner et al., 2018; Kimura et al., 2008; Sanders and Arbeitman, 2008; Taylor and Truman, 1992), including the ‘neurons medially located, just above antennal lobe’ (mAL) (Kimura et al., 2005). Normal male flies have ∼30 mAL neurons, whereas females have ∼5 mAL neurons (Kimura et al., 2005). In males, these mAL neurons extend neurite projections bilaterally, whereas the smaller number of female mAL neurons have only contralateral projections (Fig. 2A). Remarkably, sex differences in both the number and projections of mAL neurons are largely due to sex-specific programmed cell death, as genetic inhibition of apoptosis in females increases the number of mAL neurons and specifies male-like bilateral projections in ‘rescued’ mAL neurons. In the case of mAL neurons (Kimura et al., 2005) and many other neuronal populations in Drosophila (Billeter et al., 2006; Demir and Dickson, 2005; Garner et al., 2018; Kimura et al., 2008; Rideout et al., 2007, 2010; Sanders and Arbeitman, 2008; Shirangi et al., 2016; Taylor and Truman, 1992), genes in the sex determination pathway have been shown to establish and maintain this sex difference in neuron number.

Fig. 2.

Sex differences in fundamental cellular processes shape specific cell populations within organs. (A) In Drosophila melanogaster, a sex difference in programmed cell death occurs in the ‘neurons medially located, just above antennal lobe’ (mAL) (Kimura et al., 2005). In females, cell death leads to fewer mAL neurons, and surviving mAL neurons have different neurite projections from males. In combination with other male-specific neuronal populations, the sex difference in mAL neurons supports normal courtship and reproductive behaviors in males. Although this figure displays a sex difference in cell death in Drosophila neurons, sex-specific neuronal cell death occurs in many animals (e.g. worms, frogs and birds) (Forger and de Vries, 2010; Kay et al., 1999; Kirn and DeVoogd, 1989). (B) In newborn rats, there is higher proliferation in the CA1 and CA3 regions of the hippocampus in males than in females (Bowers et al., 2010; Zhang et al., 2008). This increased proliferation may contribute to the male-biased increase in neuron number in these regions of the hippocampus (Hilton et al., 2003). Although this figure displays a sex difference in proliferation in rat neurons, sex-specific proliferation occurs in the CNS of many animals (e.g. flies, worms and fish) (Ampatzis and Dermon, 2007; DeWulf and Bottjer, 2002; Ross et al., 2005; Taylor and Truman, 1992). (C) Pancreatic islets derived from human female donors contain a slightly higher proportion of insulin-producing β-cells than males; however, the fundamental cellular processes that contribute to this difference remain unknown. This modest difference in β-cell mass may contribute to the increase in glucose-stimulated insulin release that has been observed in females compared with males in the context of equivalent insulin sensitivity between the sexes (Horie et al., 2018); however, a functional link between higher β-cell mass and increased glucose-stimulated insulin secretion in females has not yet been established.

Fig. 2.

Sex differences in fundamental cellular processes shape specific cell populations within organs. (A) In Drosophila melanogaster, a sex difference in programmed cell death occurs in the ‘neurons medially located, just above antennal lobe’ (mAL) (Kimura et al., 2005). In females, cell death leads to fewer mAL neurons, and surviving mAL neurons have different neurite projections from males. In combination with other male-specific neuronal populations, the sex difference in mAL neurons supports normal courtship and reproductive behaviors in males. Although this figure displays a sex difference in cell death in Drosophila neurons, sex-specific neuronal cell death occurs in many animals (e.g. worms, frogs and birds) (Forger and de Vries, 2010; Kay et al., 1999; Kirn and DeVoogd, 1989). (B) In newborn rats, there is higher proliferation in the CA1 and CA3 regions of the hippocampus in males than in females (Bowers et al., 2010; Zhang et al., 2008). This increased proliferation may contribute to the male-biased increase in neuron number in these regions of the hippocampus (Hilton et al., 2003). Although this figure displays a sex difference in proliferation in rat neurons, sex-specific proliferation occurs in the CNS of many animals (e.g. flies, worms and fish) (Ampatzis and Dermon, 2007; DeWulf and Bottjer, 2002; Ross et al., 2005; Taylor and Truman, 1992). (C) Pancreatic islets derived from human female donors contain a slightly higher proportion of insulin-producing β-cells than males; however, the fundamental cellular processes that contribute to this difference remain unknown. This modest difference in β-cell mass may contribute to the increase in glucose-stimulated insulin release that has been observed in females compared with males in the context of equivalent insulin sensitivity between the sexes (Horie et al., 2018); however, a functional link between higher β-cell mass and increased glucose-stimulated insulin secretion in females has not yet been established.

Sex differences in cell proliferation also exist in the CNS. For example, in newborn rats, gonadal hormones contribute to a sex difference in cell proliferation in several hippocampal regions (Fig. 2B), with newborn male rats showing higher proliferation in these regions than females (Bowers et al., 2010; Zhang et al., 2008). Because there was no accompanying increase in cell death (Zhang et al., 2008), cell proliferation and not cell survival likely explains why young male rats have an increased number of neurons in these regions (Hilton et al., 2003). Indeed, sex differences in cell proliferation and death extend broadly within the CNS, occurring both during development and throughout life (Forger and de Vries, 2010). Although these differences in CNS development may seem modest, it is important to note that even small differences in cell number or dendrite projections are functionally significant.

Sex-specific neuronal clusters in several animals have been linked with diverse behaviors and physiological parameters (Barr et al., 2018; Billeter et al., 2006; Brenowitz and Arnold, 1986; Cahill, 2006; Hellier et al., 2018; Narayan et al., 2016; Nottebohm and Arnold, 1976; Pavlou et al., 2016; Portman, 2007; Rezával et al., 2014; Rideout et al., 2007; Sammut et al., 2015; Shirangi et al., 2016; Wang et al., 2018). For example, in C. elegans, one pair of male-specific MCM neurons mediates the ability of male worms, but not hermaphrodites, to undergo a switch in chemosensory behavior after exposure to a potential mate, a form of sexual conditioning (Sakai et al., 2013; Sammut et al., 2015). Similarly, 30 TN1 interneurons present in the male, but not the female, ventral nerve cord in Drosophila are required for the production of sine song, a key component of the male courtship ritual (Rideout et al., 2007; Sanders and Arbeitman, 2008; Shirangi et al., 2016). In mice, sexual dimorphism in POMC neuron number and activity contribute to the male-female difference in diet-induced obesity (Wang et al., 2018). Although the male-specific MCM and TN1 neurons in C. elegans and D. melanogaster, respectively, are produced through the action of sex determination genes (Rideout et al., 2007; Sammut et al., 2015; Shirangi et al., 2016), the mechanisms underlying the sexual dimorphism in POMC number remain unclear. Together, these findings and others demonstrate that even small differences in neuron number between the sexes can influence behavior and physiology.

These examples of sex-specific cell proliferation and death illustrate why studying both sexes helps to understand the fundamental mechanisms that shape organ and tissue properties. This is equally important in organs with and without obvious sexual dimorphism (Fig. 2C), as sex differences are found across many organs. Benefits arising from studying both sexes also apply to studies on processes in addition to cell proliferation and death, as multiple cellular mechanisms may contribute to differences in tissue and organ mass (e.g. cell proliferation, growth, death, competition and migration). Indeed, there is a significant risk of overlooking a fundamental cellular process that shapes tissue and organ size when using single- or mixed-sex populations. Furthermore, without certainty about whether a group of cells is present in both sexes or not, incorrect conclusions may be drawn about the functional significance of that cell type. Studying both sexes, and including biological sex as a variable during data analysis, will therefore allow a more accurate understanding of how fundamental cellular processes influence tissue and organ development, and of how these organs function within the body.

Knowledge of mechanisms that operate at the level of whole organs to maintain tissue and organ homeostasis is also required for a complete understanding of development. In some organs (e.g. intestine and skin), maintaining homeostasis requires cells of the correct type to be replaced in a regulated manner according to cell loss (Biteau et al., 2011; Fuchs, 2008; Jiang and Edgar, 2011). Although cell loss may occur under normal physiological conditions, it also occurs following mechanical, biological and chemical insults (e.g. injury and infection). In many animals, sex differences have been reported in both tissue and organ homeostasis, and in their capacity for repair, suggesting the mechanisms underlying these processes differ between the sexes.

Sex differences in stem cell behavior influence organ plasticity, repair and aging

One example of a sex difference in organ homeostasis and repair emerges from studies on intestinal stem cells (ISC) in the Drosophila melanogaster gut (Ahmed et al., 2020; Hudry et al., 2016; Regan et al., 2016). Normally, ISCs divide asymmetrically to produce a daughter cell called an enteroblast (EB) (Micchelli and Perrimon, 2006; Ohlstein and Spradling, 2006). EBs are transient progenitors that differentiate into enterocytes (EC) and enteroendocrine cells (EE), two important cell types in the adult intestine (Jiang and Edgar, 2012; Lemaitre and Miguel-Aliaga, 2013). ISC proliferation therefore maintains intestinal homeostasis under normal physiological conditions (Micchelli and Perrimon, 2006; Ohlstein and Spradling, 2006) and plays a crucial role in repairing damage due to mechanical and biological insults (Amcheslavsky et al., 2009; Buchon et al., 2009; Jiang et al., 2009).

Several studies now show that, under normal physiological conditions, the proliferation of female ISCs is ∼400-800% higher than the proliferation of male ISCs (Ahmed et al., 2020; Hudry et al., 2016; Regan et al., 2016). This difference exists after flies ingest a detergent to damage the intestinal epithelium (Ahmed et al., 2020; Hudry et al., 2016) and after oral infection with Erwinia carotovora or Pseudomonas entomophila (Ahmed et al., 2020; Regan et al., 2016), and persists throughout life (Regan et al., 2016). The sex difference in proliferation is due to intrinsic ISC sexual identity, and is further enhanced in females following a mating-induced increase in the steroid hormone ecdysone (Ahmed et al., 2020; Reiff et al., 2015; Zipper et al., 2020). Importantly, the sex difference in ISC proliferation has physiological consequences: higher ISC proliferation in females leads to a larger intestine (Hudry et al., 2016) and may contribute to increased female resistance to infection-related insults (Regan et al., 2016).

Despite these benefits, however, females suffer more age-related damage to the intestinal epithelium (Regan et al., 2016) and increased susceptibility to genetically induced tumors (Ahmed et al., 2020; Hudry et al., 2016). A sex difference in stem cell proliferation therefore not only has important consequences for normal physiology, but also influences the risk of disease. Although less is known in other models about sex differences in ISCs and intestinal homeostasis, a recent study documented higher ISC proliferation in female mice (Zhou et al., 2018). Future studies will need to resolve whether this difference leads to male-female differences in gut size, plasticity and capacity for repair in mammals, as it does in flies. Because the sex difference in ISC proliferation was not driven by a direct effect of estrogen, it will also be important to determine the mechanism underlying increased female ISC divisions.

An established literature on sex differences in hematopoietic stem cells (Nakada et al., 2014) similarly suggests that more work is needed to understand how stem cells contribute to male-female differences in the regulation and function of the immune system (Grossman, 1984; Klein and Flanagan, 2016). Given that sex differences also exist in adrenal stem cells (Grabek et al., 2019), neural stem cells (Bramble et al., 2019; Mahmoud et al., 2016), adipose-derived stromal cells (Ogawa et al., 2004), multipotent mesenchymal stromal cells (Bragdon et al., 2015) and muscle-derived stem cells (Deasy et al., 2007), there is currently an unmet need for knowledge of how sex differences in stem cell behavior impact organ homeostasis and repair throughout the body.

Exploiting sex differences in stem cell behavior to optimize outcomes in tissue engineering and regenerative medicine

In addition to studying tissue and organ homeostasis, sex differences in stem cell behavior also impact the production and function of stem cells for downstream applications, including regenerative medicine and tissue engineering. For example, muscle-derived stem cells isolated from female mice are more efficient at regenerating skeletal muscle than male stem cells when implanted into a mouse model of Duchenne muscular dystrophy (Fig. 3A) (Deasy et al., 2007). The increased regeneration ability of female muscle-derived stem cells is largely due to a sex difference in myogenic differentiation upon cell stress in the implantation environment, where increased post-implant differentiation in male muscle-derived stem cells potentially depletes the stem cell pool available for regeneration.

Fig. 3.

Sex differences in stem cell behavior influence tissue regeneration and organoid formation, and host-derived factors impact maturation of implanted progenitor cells. (A) Muscle-derived stem cells isolated from female donors support higher muscle regeneration in a mouse model of Duchenne muscular dystrophy (Deasy et al., 2007). (B) Colon crypts isolated from female donors produced colonoids larger in size and greater in number than colonoids generated from male crypts (Noguerol et al., 2019). (C) Stage 4 (S4) human embryonic stem cell-derived pancreatic progenitors implanted into female recipients demonstrated significant glucose responsiveness weeks earlier than S4 pancreatic progenitors implanted into males (Saber et al., 2018). As a result, blood glucose levels were lower in female mice than in males following a glucose challenge between 2 and 20 weeks post-implant, suggesting earlier maturation of S4 cells in females improved implant function.

Fig. 3.

Sex differences in stem cell behavior influence tissue regeneration and organoid formation, and host-derived factors impact maturation of implanted progenitor cells. (A) Muscle-derived stem cells isolated from female donors support higher muscle regeneration in a mouse model of Duchenne muscular dystrophy (Deasy et al., 2007). (B) Colon crypts isolated from female donors produced colonoids larger in size and greater in number than colonoids generated from male crypts (Noguerol et al., 2019). (C) Stage 4 (S4) human embryonic stem cell-derived pancreatic progenitors implanted into female recipients demonstrated significant glucose responsiveness weeks earlier than S4 pancreatic progenitors implanted into males (Saber et al., 2018). As a result, blood glucose levels were lower in female mice than in males following a glucose challenge between 2 and 20 weeks post-implant, suggesting earlier maturation of S4 cells in females improved implant function.

Another example is a sex difference in the ability of male- and female-derived stem cells to form self-organized three-dimensional organ or tissue cultures called organoids (Fig. 3B). Stem cell-derived organoids reproduce some aspects of organ or tissue structure and gene expression, and in some cases may recapitulate functional properties of the organ or tissue from which the stem cells were isolated. One recent study showed that colonoids cultured from colon crypts of females were not only more numerous when compared with males, but female colonoids also had a significantly larger size (Noguerol et al., 2019).

Beyond considerations of donor sex in stem cell studies, both sexes must also be included as recipients of stem cells and stem cell-derived tissues. The importance of this point is shown by the effect of recipient sex on the maturation of stage 4 (S4) human embryonic stem cell-derived pancreatic progenitors into glucose-responsive insulin-secreting cells (Fig. 3C) (Saber et al., 2018). S4 pancreatic progenitors mature into glucose-responsive insulin-producing cells when implanted into a host and restore glucose control when implanted in mice with impaired insulin function (Bruin et al., 2013; Rezania et al., 2012). Interestingly, S4 pancreatic progenitors implanted into females demonstrated significant glucose responsiveness weeks earlier than pancreatic progenitors implanted into males (Saber et al., 2018). Because glucose responsiveness is an important attribute of pancreatic progenitors intended for therapeutic purposes, it is biologically significant that progenitors implanted into females achieve this goal sooner.

Together, these studies highlight why both sexes must be included when investigating how stem cell behavior influences organ homeostasis and repair. This applies to stem cell studies in the context of living animals and also to studies aimed at using stem cells for applications such as regenerative medicine or tissue engineering. When both sexes are not studied, there is a risk of over- or underestimating the capacity of an organ for repair and an individual's ability to survive loss of organ function. Conversely, accounting for sex provides benefits such as identifying organoids with the most desirable characteristics, and uncovering the stem cells most likely to regenerate lost or damaged tissue. In addition, when stem cells are implanted into both male and female recipients, the functional properties of implanted cells within a host will be described with more accuracy.

A complete mechanistic understanding of development also requires knowledge of mechanisms that operate at the level of the whole organism to control whole-body phenotypes. In all animals, signals sent between tissues and organs, such as growth factors and hormones, play a key role in regulating development at the level of the whole organism (Leopold and Perrimon, 2007). These signals may be released locally to act upon neighboring cells, or into the circulation to impact distant cells. On target cells, these signals interact with receptors to activate cell signaling pathways, triggering an intracellular response. The coordinated activation of cell signaling pathways in multiple tissues and organs supports development at the whole-organism level. Importantly, sex differences in cell signaling pathways and in phenotypes regulated at the organismal level have been detected in multiple animals (Millington and Rideout, 2018).

Sex differences in signaling pathways affect development under normal physiological conditions

One example of how sex differences in cell signaling pathways affect development is the insulin/insulin-like growth factor signaling pathway (IIS). In all animals, IIS plays a key role in regulating growth during development (Grewal, 2009; Oldham and Hafen, 2003; Teleman, 2010). In both invertebrates and vertebrates, binding of IIS ligands to a receptor triggers the activation of an intracellular signaling cascade in target tissues to augment cell and tissue growth, leading to a larger body size. IIS activity is therefore a key determinant of final body size across multiple species. Notably, studies in diverse animals report male-female differences in IIS regulation and function.

In Drosophila melanogaster, IIS activity is higher in late third-instar female larvae than in age-matched males (Rideout et al., 2015; Millington et al., 2021a). Several factors likely contribute to increased IIS activity in females, including increased secretion of Drosophila insulin-like peptide 2 (Dilp2) (Rideout et al., 2015). Females also have higher levels of factors that stimulate Dilp2 secretion (Millington et al., 2021a) and lower levels of proteins that interact with Dilp2 to inhibit binding with the insulin receptor (Arquier et al., 2008; Honegger et al., 2008; Millington et al., 2021b). Thus, in Drosophila larvae, there is a robust difference in IIS activity between the sexes due to sex-specific regulation of IIS ligands and factors that influence ligand-receptor interactions. Although the mechanisms underlying this sex-specific IIS regulation remain incompletely understood, the function of the sex determination gene transformer contributes to the male-female difference in Dilp2 secretion (Millington et al., 2021a; Rideout et al., 2015).

In mammals, it is difficult to generalize about a sex difference in IIS activity due to multiple receptors and tissue-specific requirements for IIS in regulating body size; however, sex differences in IIS ligands have been reported (Gatford et al., 1998). In rats, mice, pigs, sheep and cattle, levels of the IIS ligand insulin-like growth factor 1 (IGF1) are higher in males than in females, although IGF1 levels are higher in female primates (Gatford et al., 1998). Circulating levels of another IIS ligand, insulin, are also higher in female humans than in males (Basu et al., 2006, 2017), whereas a sex difference in IGF2 levels remains unclear (Geary et al., 2003; Yu et al., 1999). As in flies, sex differences in IGF-binding and -interacting proteins have also been observed (Geary et al., 2003; Yu et al., 1999), suggesting sex-specific IIS regulation exists across diverse species.

Although IIS activity is only 20% higher in Drosophila females compared with males (Rideout et al., 2015; Millington et al., 2021a), this difference is physiologically significant: adult females are normally 30% larger than adult males, a difference that is strongly reduced or abolished upon decreased IIS activity (Testa et al., 2013; Millington et al., 2021b). In mammals, although total IGF1 levels are only modestly higher in males than in females, this difference allows males to achieve a heavier body weight than females (Ashpole et al., 2017; Baker et al., 1993; Bikle et al., 2001; Liu et al., 1993; Powell-Braxton et al., 1993). Thus, even minor differences in cell signaling activity between males and females cause visible differences in development.

Although we focused on the sex-specific regulation and function of IIS in this section in order to illuminate many ways in which a pathway may differ between the sexes, it is essential to note that multiple pathways have been implicated in regulating male-female differences in development. For example, studies have identified sex-specific requirements for different ligands of the bone morphogenetic protein (BMP) signaling pathway in regulating gametogenesis and reproductive processes in many animals (reviewed by Lochab and Extavour, 2017; Shah and Rogers, 2018), and sex hormones regulate BMP family members across many cell types (reviewed by Shah and Rogers, 2018). Similarly, male-specific production of a janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway ligand by the somatic gonad early in development activates pathway activity in the germline to promote male germ cell behavior (Wawersik et al., 2005). Indeed, there are clues from multiple animals that many signaling pathways (e.g. Notch, Hedgehog and Wnt) are sex-specifically regulated during gonad development (DeFalco et al., 2003; Deshpande et al., 2016; Franco and Yao, 2012; Liu et al., 2009; Penn and Schedl, 2007; Sanchez et al., 2001; Windley and Wilhelm, 2016), findings that may be relevant for other aspects of development and physiology regulated by these important pathways (e.g. stem cell proliferation, metabolic regulation and body size).

Sex differences in cell signaling pathways in response to environmental and life history changes

Changes in environmental conditions (e.g. temperature and nutrient availability) and life history (e.g. reproduction) also modulate the activity of specific cell signaling pathways. These context-dependent changes to cell signaling stimulate metabolic and physiological changes required to maintain homeostasis, to survive and to reproduce. The ability of an individual to coordinate pathway activity with environmental cues or life history changes is therefore an important determinant of stress resistance, survival and reproduction. Studies in multiple contexts now show that males and females differ in the coordination of cell signaling activity with environmental stimuli and life history events. For example, in rhinoceros beetles, there is sex-limited coupling between nutrients and horn size (Emlen et al., 2012). Normally, male beetles have horns and females do not. In nutrient-rich conditions, male beetle horns grow significantly larger, whereas a nutrient-poor diet causes the development of smaller horns. Female beetles, in contrast, do not show any nutrient-dependent changes in horn size. Interestingly, these effects on horn size were mediated by male-specific changes to IIS activity, demonstrating sex-limited coupling between an environmental condition and IIS regulation.

Similar sex-biased coupling between nutrients and cell signaling pathways has been reported in Drosophila melanogaster, where females have an increased ability compared with males to adjust body and trait size in response to several nutrients (Shingleton et al., 2017; McDonald et al., 2020; Millington et al., 2021a). In the case of dietary protein, female larvae reared on a protein-rich diet display a greater increase in body size than males (Millington et al., 2021a). Interestingly, the protein-rich diet triggers a female-biased increase in IIS activity, suggesting that IIS activity is more tightly coupled with protein intake in females than in males (Millington et al., 2021a). This sex-biased coupling between environmental cues and cell signaling extends to the activation of the Toll pathway following infection (Duneau et al., 2017), activation of cellular responses to oxidative stress in Drosophila (Pomatto et al., 2017) and to neurons that mediate behavioral responses to chemosensory cues (e.g. diacetyl and sodium dodecyl sulphate) in C. elegans (Bayer et al., 2020; Lawson et al., 2020; Ryan et al., 2014). Because similar differences have been recorded with other signaling pathways and animals (Baar et al., 2016; Clegg et al., 2003; Duneau et al., 2017; Havel et al., 1996; Klein and Flanagan, 2016; Saad et al., 1997; Testa and Dworkin, 2016; Tower et al., 2020), sex differences in the coupling between environmental factors and cell signaling pathways are found across many species. Beyond sex-specific coordination between environmental cues and cell signaling, sex-specific life history events contribute to male-female differences in cell signaling. This is exemplified by mating-dependent changes to female physiology and behavior (Avila et al., 2011; Chapman, 2001; Gillott, 2003; Pitnick et al., 2009; Poiani, 2006; Rodríguez-Martínez et al., 2011; Wolfner, 1997; Adams and Wolfner, 2007; Ameku and Niwa, 2016; Apger-MdGlaughon and Wolfner, 2013; Avila and Wolfner, 2009; Carvalho et al., 2006; Cognigni et al., 2011; Hadjieconomou et al., 2020; Harshman et al., 1999; Heifets and Wolfner, 2004; Mattei et al., 2015; Rubinstein and Wolfner, 2013; Sieber and Spradling, 2015; Vargas et al., 2010; Ribeiro and Dickson, 2010; Walker et al., 2015). Importantly, these mating-induced phenotypes have been shown to depend on specific hormonal and cell signaling pathways, indicating that a sex difference in life history events such as mating can be an important way to achieve a sex difference in cell signaling.

Sex differences in cell signaling are often subtle; however, there are important consequences for development, metabolic regulation, survival and lifespan. For example, although changes to nutrient quantity and IIS activity in Drosophila females significantly influence body size and lifespan, nutrient- and IIS-dependent changes to these phenotypes are less severe or absent in males (Clancy et al., 2001; Giannakou, 2004; Magwere et al., 2004; Millington et al., 2021a,b; Tatar et al., 2001; Woodling et al., 2020). Changes to nutrients and IIS activity often, but not always, also have female-biased effects on lifespan in other animals (Bokov et al., 2011; Holzenberger et al., 2003; Honjoh et al., 2017; Mao et al., 2018; Mitchell et al., 2016; Templeman et al., 2017; Xu et al., 2014).

When taken together, these studies highlight how including both sexes in studies on cell signaling pathways provides an opportunity to gain a deeper mechanistic understanding of how these pathways affect development. This principle holds when studying development in a normal physiological context, and when investigating how development is affected by diverse environmental conditions and life history events. Using a single- or mixed-sex population risks overlooking profound phenotypic consequences caused by changes to cell signaling activity if the affected sex does not comprise a large enough proportion of the experimental population. Similarly, there is a risk of making inaccurate predictions about the resilience of each sex to environmental variation, and about which pathways mediate these effects (Noiret et al., 2020).

Sex differences in developmental mechanisms are found across species and at each level of biological complexity. Although it is easy to predict that different mechanisms will operate in males and females for traits with visible differences between the sexes, distinct mechanisms also operate in the large number of organs with less conspicuous differences. Because we are likely aware of only a minority of sex differences within organs due to extensive use of single- and mixed-sex animal groups, as a community, we should normalize the inclusion of males and females in developmental biology studies. Successful and widespread inclusion of both sexes will provide several important benefits. First, we will gain a more-complete and accurate understanding of the genetic and molecular mechanisms underlying many aspects of development, behavior and physiology. When both sexes are not studied separately, the contribution of a specific gene to development may be obscured in mixed-sex groups or completely overlooked in single-sex groups. Second, we will gain a better understanding of the fundamental mechanisms that govern tissue and organ size control and the capacity of tissues and organs for repair. This is essential, as both size and capacity for repair are related to how well an organ performs its vital functions within the body. Third, we will improve our understanding of the cell signaling pathways that regulate development and maintain homeostasis in each sex. Given that signaling pathways regulate multiple aspects of development, influence organismal phenotypes (e.g. stress resistance, successful reproduction, lifespan) and contribute to disease, including males and females in developmental biology studies will provide insights relevant for a very broad scientific community.

We thank Jacelyn Shu (www.jacelyndesigns.com) for drafting Fig. 1. We also thank Rideout lab members for thoughtful comments on the manuscript. We acknowledge that our research takes place on the traditional, ancestral and unceded territory of the Musqueam people; a privilege for which we are grateful.

Funding

E.J.R. acknowledges funding from the Canadian Institutes for Health Research (CIHR, PJT-153072), the Canadian Institutes for Health Research Sex and Gender Science Chair Program (GS4-171365), the Natural Sciences and Engineering Research Council of Canada (NSERC, RGPIN-2016-04249), the Michael Smith Foundation for Health Research (16876), and the Canadian Foundation for Innovation (JELF-34879). J.E.M. acknowledges funding from the Canada 150 Research Chair program, the Natural Sciences and Engineering Research Council of Canada and the European Research Council (grant number 680951).

Adams
,
E. M.
and
Wolfner
,
M. F.
(
2007
).
Seminal proteins but not sperm induce morphological changes in the Drosophila melanogaster female reproductive tract during sperm storage
.
J. Insect Physiol.
53
,
319
-
331
.
Ahmed
,
S. M. H.
,
Maldera
,
J. A.
,
Krunic
,
D.
,
Paiva-Silva
,
G. O.
,
Pénalva
,
C.
,
Teleman
,
A. A.
and
Edgar
,
B. A.
(
2020
).
Fitness trade-offs incurred by ovary-to-gut steroid signalling in Drosophila
.
Nature
584
,
415
-
419
.
Amcheslavsky
,
A.
,
Jiang
,
J.
and
Ip
,
Y. T.
(
2009
).
Tissue damage-induced intestinal stem cell division in Drosophila
.
Cell Stem Cell
4
,
49
-
61
.
Ameku
,
T.
and
Niwa
,
R.
(
2016
).
Mating-induced increase in germline stem cells via the neuroendocrine system in female Drosophila
.
PLoS Genet.
12
,
e1006123
.
Ampatzis
,
K.
and
Dermon
,
C. R.
(
2007
).
Sex differences in adult cell proliferation within the zebrafish (Danio rerio) cerebellum
.
Eur. J. Neurosci.
25
,
1030
-
1040
.
Apger-McGlaughon
,
J.
and
Wolfner
,
M. F.
(
2013
).
Post-mating change in excretion by mated Drosophila melanogaster females is a long-term response that depends on sex peptide and sperm
.
J. Insect Physiol.
59
,
1024
-
1030
.
Ardia
,
D. R.
,
Broughton
,
D. R.
and
Gleicher
,
M. J.
(
2010
).
Short-term exposure to testosterone propionate leads to rapid bill color and dominance changes in zebra finches
.
Horm. Behav.
58
,
526
-
532
.
Arnold
,
A. P.
(
2017
).
A general theory of sexual differentiation
.
J. Neurosci. Res.
95
,
291
-
300
.
Arnold
,
A. P.
(
2019
).
Rethinking sex determination of non-gonadal tissues
.
Curr. Top. Dev. Biol.
134
,
289
-
315
.
Arquier
,
N.
,
Géminard
,
C.
,
Bourouis
,
M.
,
Jarretou
,
G.
,
Honegger
,
B.
,
Paix
,
A.
and
Léopold
,
P.
(
2008
).
Drosophila ALS regulates growth and metabolism through functional interaction with insulin-like peptides
.
Cell Metab.
7
,
333
-
338
.
Ashpole
,
N. M.
,
Logan
,
S.
,
Yabluchanskiy
,
A.
,
Mitschelen
,
M. C.
,
Yan
,
H.
,
Farley
,
J. A.
,
Hodges
,
E. L.
,
Ungvari
,
Z.
,
Csiszar
,
A.
,
Chen
,
S.
et al. 
(
2017
).
IGF-1 has sexually dimorphic, pleiotropic, and time-dependent effects on healthspan, pathology, and lifespan
.
GeroScience
39
,
129
-
145
.
Avila
,
F. W.
and
Wolfner
,
M. F.
(
2009
).
Acp36DE is required for uterine conformational changes in mated Drosophila females
.
Proc. Natl. Acad. Sci. USA
106
,
15796
-
15800
.
Avila
,
F. W.
,
Sirot
,
L. K.
,
LaFlamme
,
B. A.
,
Rubinstein
,
C. D.
and
Wolfner
,
M. F.
(
2011
).
Insect seminal fluid proteins: identification and function
.
Annu. Rev. Entomol.
56
,
21
-
40
.
Baar
,
E. L.
,
Carbajal
,
K. A.
,
Ong
,
I. M.
and
Lamming
,
D. W.
(
2016
).
Sex- and tissue-specific changes in mTOR signaling with age in C57BL/6J mice
.
Aging Cell
15
,
155
-
166
.
Bachtrog
,
D.
,
Mank
,
J. E.
,
Peichel
,
C. L.
,
Kirkpatrick
,
M.
,
Otto
,
S. P.
,
Ashman
,
T.-L.
,
Hahn
,
M. W.
,
Kitano
,
J.
,
Mayrose
,
I.
,
Ming
,
R.
et al. 
(
2014
).
Sex determination: why so many ways of doing it?
PLoS Biol.
12
,
e1001899
.
Badyaev
,
A. V.
(
2002
).
Growing apart: an ontogenetic perspective on the evolution of sexual size dimorphism
.
Trends Ecol. Evol.
17
,
369
-
378
.
Baker
,
J.
,
Liu
,
J.-P.
,
Robertson
,
E. J.
and
Efstratiadis
,
A.
(
1993
).
Role of insulin-like growth factors in embryonic and postnatal growth
.
Cell
75
,
73
-
82
.
Balaton
,
B. P.
and
Brown
,
C. J.
(
2016
).
Escape Artists of the X Chromosome
.
Trends Genet.
32
,
348
-
359
.
Barr
,
M. M.
,
García
,
L. R.
and
Portman
,
D. S.
(
2018
).
Sexual dimorphism and sex differences in caenorhabditis elegans neuronal development and behavior
.
Genetics
208
,
909
-
935
.
Basu
,
R.
,
Dalla Man
,
C.
,
Campioni
,
M.
,
Basu
,
A.
,
Klee
,
G.
,
Toffolo
,
G.
,
Cobelli
,
C.
and
Rizza
,
R. A.
(
2006
).
Effects of age and sex on postprandial glucose metabolism: differences in glucose turnover, insulin secretion, insulin action, and hepatic insulin extraction
.
Diabetes
55
,
2001
-
2014
.
Basu
,
A.
,
Dube
,
S.
and
Basu
,
R.
(
2017
).
Men are from mars, women are from venus: sex differences in insulin action and secretion
.
Adv. Exp. Med. Biol.
1043
,
53
-
64
.
Bayer
,
E. A.
,
Stecky
,
R. C.
,
Neal
,
L.
,
Katsamba
,
P. S.
,
Ahlsen
,
G.
,
Balaji
,
V.
,
Hoppe
,
T.
,
Shapiro
,
L.
,
Oren-Suissa
,
M.
and
Hobert
,
O.
(
2020
).
Ubiquitin-dependent regulation of a conserved DMRT protein controls sexually dimorphic synaptic connectivity and behavior
.
eLife
9
,
e59614
.
Bikle
,
D.
,
Majumdar
,
S.
,
Laib
,
A.
,
Powell-Braxton
,
L.
,
Rosen
,
C.
,
Beamer
,
W.
,
Nauman
,
E.
,
Leary
,
C.
and
Halloran
,
B.
(
2001
).
The skeletal structure of insulin-like growth factor I-deficient mice
.
J. Bone Miner Res.
16
,
2320
-
2329
.
Billeter
,
J.-C.
,
Villella
,
A.
,
Allendorfer
,
J. B.
,
Dornan
,
A. J.
,
Richardson
,
M.
,
Gailey
,
D. A.
and
Goodwin
,
S. F.
(
2006
).
Isoform-specific control of male neuronal differentiation and behavior in Drosophila by the fruitless gene
.
Curr. Biol.
16
,
1063
-
1076
.
Biteau
,
B.
,
Hochmuth
,
C. E.
and
Jasper
,
H.
(
2011
).
Maintaining tissue homeostasis: dynamic control of somatic stem cell activity
.
Cell Stem Cell
9
,
402
-
411
.
Bogetto
,
F.
,
Venturello
,
S.
,
Albert
,
U.
,
Maina
,
G.
and
Ravizza
,
L.
(
1999
).
Gender-related clinical differences in obsessive-compulsive disorder
.
Eur. Psychiatry
14
,
434
-
441
.
Bokov
,
A. F.
,
Garg
,
N.
,
Ikeno
,
Y.
,
Thakur
,
S.
,
Musi
,
N.
,
DeFronzo
,
R. A.
,
Zhang
,
N.
,
Erickson
,
R. C.
,
Gelfond
,
J.
,
Hubbard
,
G. B.
et al. 
(
2011
).
Does reduced IGF-1R signaling in Igf1r+/− mice alter aging?
PLoS ONE
6
,
e26891
.
Boraska
,
V.
,
Jerončić
,
A.
,
Colonna
,
V.
,
Southam
,
L.
,
Nyholt
,
D. R.
,
Rayner
,
N. W.
,
Perry
,
J. R. B.
,
Toniolo
,
D.
,
Albrecht
,
E.
,
Ang
,
W.
et al. 
(
2012
).
Genome-wide meta-analysis of common variant differences between men and women
.
Hum. Mol. Genet.
21
,
4805
-
4815
.
Bowers
,
J. M.
,
Waddell
,
J.
and
McCarthy
,
M. M. A.
(
2010
).
A developmental sex difference in hippocampal neurogenesis is mediated by endogenous oestradiol
.
Biol. Sex Differ.
1
,
8
.
Bragdon
,
B.
,
Burns
,
R.
,
Baker
,
A. H.
,
Belkina
,
A. C.
,
Morgan
,
E. F.
,
Denis
,
G. V.
,
Gerstenfeld
,
L. C.
and
Schlezinger
,
J. J.
(
2015
).
Intrinsic sex-linked variations in osteogenic and adipogenic differentiation potential of bone marrow multipotent stromal cells: age- and sex-linked variation in MSC differentiation
.
J. Cell. Physiol.
230
,
296
-
307
.
Bramble
,
M. S.
,
Vashist
,
N.
and
Vilain
,
E.
(
2019
).
Sex steroid hormone modulation of neural stem cells: a critical review
.
Biol. Sex Differ.
10
,
28
.
Brenowitz
,
E. A.
and
Arnold
,
A. P.
(
1986
).
Interspecific comparisons of the size of neural song control regions and song complexity in duetting birds: evolutionary implications
.
J. Neurosci.
6
,
2875
-
2879
.
Bruin
,
J. E.
,
Rezania
,
A.
,
Xu
,
J.
,
Narayan
,
K.
,
Fox
,
J. K.
,
O'Neil
,
J. J.
and
Kieffer
,
T. J.
(
2013
).
Maturation and function of human embryonic stem cell-derived pancreatic progenitors in macroencapsulation devices following transplant into mice
.
Diabetologia
56
,
1987
-
1998
.
Buchon
,
N.
,
Broderick
,
N. A.
,
Poidevin
,
M.
,
Pradervand
,
S.
and
Lemaitre
,
B.
(
2009
).
Drosophila intestinal response to bacterial infection: activation of host defense and stem cell proliferation
.
Cell Host Microbe
5
,
200
-
211
.
Cahill
,
L.
(
2006
).
Why sex matters for neuroscience
.
Nat. Rev. Neurosci.
7
,
477
-
484
.
Camus
,
M. F.
,
Piper
,
M. D.
and
Reuter
,
M.
(
2019
).
Sex-specific transcriptomic responses to changes in the nutritional environment
.
eLife
8
,
e47262
.
Capel
,
B.
(
2000
).
The battle of the sexes
.
Mech. Dev.
92
,
89
-
103
.
Carvalho
,
G. B.
,
Kapahi
,
P.
,
Anderson
,
D. J.
and
Benzer
,
S.
(
2006
).
Allocrine modulation of feeding behavior by the sex peptide of Drosophila
.
Curr. Biol.
16
,
692
-
696
.
Chapman
,
T.
(
2001
).
Seminal fluid-mediated fitness traits in Drosophila
.
Heredity
87
,
511
-
521
.
Clancy
,
D. J.
,
Gems
,
D.
,
Harshman
,
L. G.
,
Oldham
,
S.
,
Stocker
,
H.
,
Hafen
,
E.
,
Leevers
,
S. J.
and
Partridge
,
L.
(
2001
).
Extension of life-span by loss of CHICO, a Drosophila insulin receptor substrate protein
.
Science
292
,
104
-
106
.
Clayton
,
J. A.
and
Collins
,
F. S.
(
2014
).
Policy: NIH to balance sex in cell and animal studies
.
Nature
509
,
282
-
283
.
Clegg
,
D. J.
,
Riedy
,
C. A.
,
Smith
,
K. A. B.
,
Benoit
,
S. C.
and
Woods
,
S. C.
(
2003
).
Differential sensitivity to central leptin and insulin in male and female rats
.
Diabetes
52
,
682
-
687
.
Clocchiatti
,
A.
,
Cora
,
E.
,
Zhang
,
Y.
and
Dotto
,
G. P.
(
2016
).
Sexual dimorphism in cancer
.
Nat. Rev. Cancer
16
,
330
-
339
.
Cognigni
,
P.
,
Bailey
,
A. P.
and
Miguel-Aliaga
,
I.
(
2011
).
Enteric neurons and systemic signals couple nutritional and reproductive status with intestinal homeostasis
.
Cell Metab.
13
,
92
-
104
.
Connallon
,
T.
and
Clark
,
A. G.
(
2011
).
Association between sex-biased gene expression and mutations with sex-specific phenotypic consequences in Drosophila
.
Genome Biol. Evol.
3
,
151
-
155
.
Cox
,
R. M.
,
Skelly
,
S. L.
,
Leo
,
A.
and
John-Alder
,
H. B.
(
2005
).
Testosterone regulates sexually dimorphic coloration in the eastern fence lizard, sceloporus undulatus
.
Copeia
2005
,
597
-
608
.
de Simone
,
G.
,
Devereux
,
R. B.
,
Daniels
,
S. R.
,
Koren
,
M. J.
,
Meyer
,
R. A.
and
Laragh
,
J. H.
(
1995
).
Effect of growth on variability of left ventricular mass: assessment of allometric signals in adults and children and their capacity to predict cardiovascular risk
.
J. Am. Coll. Cardiol.
25
,
1056
-
1062
.
Deasy
,
B. M.
,
Lu
,
A.
,
Tebbets
,
J. C.
,
Feduska
,
J. M.
,
Schugar
,
R. C.
,
Pollett
,
J. B.
,
Sun
,
B.
,
Urish
,
K. L.
,
Gharaibeh
,
B. M.
,
Cao
,
B.
et al. 
(
2007
).
A role for cell sex in stem cell-mediated skeletal muscle regeneration: female cells have higher muscle regeneration efficiency
.
J. Cell Biol.
177
,
73
-
86
.
DeFalco
,
T. J.
,
Verney
,
G.
,
Jenkins
,
A. B.
,
McCaffery
,
J. M.
,
Russell
,
S.
and
Van Doren
,
M.
(
2003
).
Sex-specific apoptosis regulates sexual dimorphism in the Drosophila embryonic gonad
.
Dev. Cell
5
,
205
-
216
.
Demir
,
E.
and
Dickson
,
B. J.
(
2005
).
fruitless splicing specifies male courtship behavior in Drosophila
.
Cell
121
,
785
-
794
.
Deng
,
X.
,
Berletch
,
J. B.
,
Nguyen
,
D. K.
and
Disteche
,
C. M.
(
2014
).
X chromosome regulation: diverse patterns in development, tissues and disease
.
Nat. Rev. Genet.
15
,
367
-
378
.
Deshpande
,
G.
,
Nouri
,
A.
and
Schedl
,
P.
(
2016
).
Wnt signaling in sexual dimorphism
.
Genetics
202
,
661
-
673
.
DeWulf
,
V.
and
Bottjer
,
S. W.
(
2002
).
Age and sex differences in mitotic activity within the zebra finch telencephalon
.
J. Neurosci.
22
,
4080
-
4094
.
Dickinson
,
M. E.
,
Flenniken
,
A. M.
,
Ji
,
X.
,
Teboul
,
L.
,
Wong
,
M. D.
,
White
,
J. K.
,
Meehan
,
T. F.
,
Weninger
,
W. J.
,
Westerberg
,
H.
,
Adissu
,
H.
et al. 
(
2016
).
High-throughput discovery of novel developmental phenotypes
.
Nature
537
,
508
-
514
.
Duncan
,
L. E.
,
Ratanatharathorn
,
A.
,
Aiello
,
A. E.
,
Almli
,
L. M.
,
Amstadter
,
A. B.
,
Ashley-Koch
,
A. E.
,
Baker
,
D. G.
,
Beckham
,
J. C.
,
Bierut
,
L. J.
,
Bisson
,
J.
et al. 
(
2018
).
Largest GWAS of PTSD (N=20070) yields genetic overlap with schizophrenia and sex differences in heritability
.
Mol. Psychiatry
23
,
666
-
673
.
Duneau
,
D. F.
,
Kondolf
,
H. C.
,
Im
,
J. H.
,
Ortiz
,
G. A.
,
Chow
,
C.
,
Fox
,
M. A.
,
Eugénio
,
A. T.
,
Revah
,
J.
,
Buchon
,
N.
and
Lazzaro
,
B. P.
(
2017
).
The toll pathway underlies host sexual dimorphism in resistance to both Gram-negative and Gram-positive bacteria in mated Drosophila
.
BMC Biol.
15
,
124
.
Ecelbarger
,
C. M.
(
2016
).
Sex differences in renal physiology and pathophysiology
. In
Sex Differences in Physiology
(ed.
G.
Neigh
and
M.
Mitzelfelt
), pp.
105
-
124
,
Elsevier
.
Emlen
,
D. J.
,
Warren
,
I. A.
,
Johns
,
A.
,
Dworkin
,
I.
and
Lavine
,
L. C.
(
2012
).
A mechanism of extreme growth and reliable signaling in sexually selected ornaments and weapons
.
Science
337
,
860
-
864
.
Forger
,
N. G.
and
de Vries
,
G. J.
(
2010
).
Cell death and sexual differentiation of behavior: worms, flies, and mammals
.
Curr. Opin. Neurobiol.
20
,
776
-
783
.
Franco
,
H. L.
and
Yao
,
H. H.-C.
(
2012
).
Sex and hedgehog: roles of genes in the hedgehog signaling pathway in mammalian sexual differentiation
.
Chromosome Res.
20
,
247
-
258
.
Fuchs
,
E.
(
2008
).
Skin stem cells: rising to the surface
.
J. Cell Biol.
180
,
273
-
284
.
Galouzis
,
C. C.
and
Prud'homme
,
B.
(
2021
).
Transvection regulates the sex-biased expression of a fly X-linked gene
.
Science
371
,
396
-
400
.
Gannon
,
M.
,
Kulkarni
,
R. N.
,
Tse
,
H. M.
and
Mauvais-Jarvis
,
F.
(
2018
).
Sex differences underlying pancreatic islet biology and its dysfunction
.
Mol. Metab.
15
,
82
-
91
.
Garner
,
S. R. C.
,
Castellanos
,
M. C.
,
Baillie
,
K. E.
,
Lian
,
T.
and
Allan
,
D. W.
(
2018
).
Drosophila female-specific Ilp7 motoneurons are generated by Fruitless-dependent cell death in males and by a double-assurance survival role for transformer in females
.
Development
145
,
dev150821
.
Gatford
,
K. L.
,
Egan
,
A. R.
,
Clarke
,
I. J.
and
Owens
,
P. C.
(
1998
).
Sexual dimorphism of the somatotrophic axis
.
J. Endocrinol.
157
,
373
-
389
.
Ge
,
T.
,
Chen
,
C.-Y.
,
Neale
,
B. M.
,
Sabuncu
,
M. R.
and
Smoller
,
J. W.
(
2017
).
Phenome-wide heritability analysis of the UK Biobank
.
PLoS Genet.
13
,
e1006711
.
Geary
,
M. P. P.
,
Pringle
,
P. J.
,
Rodeck
,
C. H.
,
Kingdom
,
J. C. P.
and
Hindmarsh
,
P. C.
(
2003
).
Sexual dimorphism in the growth hormone and insulin-like growth factor axis at birth
.
J. Clin. Endocrinol. Metab.
88
,
3708
-
3714
.
Giannakou
,
M. E.
(
2004
).
Long-lived Drosophila with overexpressed dFOXO in adult fat body
.
Science
305
,
361
.
Gillott
,
C.
(
2003
).
Male accessory gland secretions: modulators of female reproductive physiology and behavior
.
Annu. Rev. Entomol.
48
,
163
-
184
.
Gordon
,
S. P.
,
López-Sepulcre
,
A.
and
Reznick
,
D. N.
(
2012
).
Predation-associated differences in sex linkage of wild guppy coloration
.
Evolution
66
,
912
-
918
.
Grabek
,
A.
,
Dolfi
,
B.
,
Klein
,
B.
,
Jian-Motamedi
,
F.
,
Chaboissier
,
M.-C.
and
Schedl
,
A.
(
2019
).
The adult adrenal cortex undergoes rapid tissue renewal in a sex-specific manner
.
Cell Stem Cell
25
,
290
-
296.e2
.
Graham
,
S. E.
,
Nielsen
,
J. B.
,
Zawistowski
,
M.
,
Zhou
,
W.
,
Fritsche
,
L. G.
,
Gabrielsen
,
M. E.
,
Skogholt
,
A. H.
,
Surakka
,
I.
,
Hornsby
,
W. E.
,
Fermin
,
D.
et al. 
(
2019
).
Sex-specific and pleiotropic effects underlying kidney function identified from GWAS meta-analysis
.
Nat. Commun.
10
,
1847
.
Graze
,
R. M.
,
Tzeng
,
R.-Y.
,
Howard
,
T. S.
and
Arbeitman
,
M. N.
(
2018
).
Perturbation of IIS/TOR signaling alters the landscape of sex-differential gene expression in Drosophila
.
BMC Genomics
19
,
893
.
Greenblatt
,
D. J.
,
Harmatz
,
J. S.
,
von Moltke
,
L. L.
,
Wright
,
C. E.
,
Durol
,
A. L.
,
Harrel-Joseph
,
L. M.
and
Shader
,
R. I.
(
2000
).
Comparative kinetics and response to the benzodiazepine agonists triazolam and zolpidem: evaluation of sex-dependent differences
.
J. Pharmacol. Exp. Ther.
293
,
435
-
443
.
Grewal
,
S. S.
(
2009
).
Insulin/TOR signaling in growth and homeostasis: a view from the fly world
.
Int. J. Biochem. Cell Biol.
41
,
1006
-
1010
.
Grossman
,
C. J.
(
1984
).
Regulation of the immune system by sex steroids*
.
Endocrine Rev.
5
,
435
-
455
.
Haast
,
R. A. M.
,
Gustafson
,
D. R.
and
Kiliaan
,
A. J.
(
2012
).
Sex differences in stroke
.
J. Cereb. Blood Flow Metab.
32
,
2100
-
2107
.
Hadjieconomou
,
D.
,
King
,
G.
,
Gaspar
,
P.
,
Mineo
,
A.
,
Blackie
,
L.
,
Ameku
,
T.
,
Studd
,
C.
,
de Mendoza
,
A.
,
Diao
,
F.
,
White
,
B. H.
et al. 
(
2020
).
Enteric neurons increase maternal food intake during reproduction
.
Nature
587
,
455
-
459
.
Harper
,
J. A.
,
Janicke
,
T.
and
Morrow
,
E. H.
(
2020
).
Systematic review reveals multiple sexually antagonistic polymorphisms affecting human disease and complex traits
.
medRxiv
2020.12.16.20248300
.
Harshman
,
L. G.
,
Loeb
,
A. M.
and
Johnson
,
B. A.
(
1999
).
Ecdysteroid titers in mated and unmated Drosophila melanogaster females
.
J. Insect. Physiol.
45
,
571
-
577
.
Havel
,
P. J.
,
Kasim-karakas
,
S.
,
Dubuc
,
G. R.
,
Muller
,
W.
and
Phinney
,
S. D.
(
1996
).
Gender differences in plasma leptin concentrations
.
Nat. Med.
2
,
949
-
950
.
Heid
,
I. M.
,
Jackson
,
A. U.
,
Randall
,
J. C.
,
Winkler
,
T. W.
,
Qi
,
L.
,
Steinthorsdottir
,
V.
,
Thorleifsson
,
G.
,
Zillikens
,
M. C.
,
Speliotes
,
E. K.
,
Mägi
,
R.
et al. 
(
2010
).
Meta-analysis identifies 13 new loci associated with waist-hip ratio and reveals sexual dimorphism in the genetic basis of fat distribution
.
Nat. Genet.
42
,
949
-
960
.
Heifetz
,
Y.
and
Wolfner
,
M. F.
(
2004
).
Mating, seminal fluid components, and sperm cause changes in vesicle release in the Drosophila female reproductive tract
.
Proc. Natl. Acad. Sci. USA
101
,
6261
-
6266
.
Hellier
,
V.
,
Brock
,
O.
,
Candlish
,
M.
,
Desroziers
,
E.
,
Aoki
,
M.
,
Mayer
,
C.
,
Piet
,
R.
,
Herbison
,
A.
,
Colledge
,
W. H.
,
Prévot
,
V.
et al. 
(
2018
).
Female sexual behavior in mice is controlled by kisspeptin neurons
.
Nat. Commun.
9
,
400
.
Hilton
,
G. D.
,
Nuez
,
J. L.
and
McCarthy
,
M. M.
(
2003
).
Sex differences in response to kainic acid and estradiol in the hippocampus of newborn rats
.
Neuroscience
116
,
383
-
391
.
Holzenberger
,
M.
,
Dupont
,
J.
,
Ducos
,
B.
,
Leneuve
,
P.
,
Géloën
,
A.
,
Even
,
P. C.
,
Cervera
,
P.
and
Le Bouc
,
Y.
(
2003
).
IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice
.
Nature
421
,
182
-
187
.
Honegger
,
B.
,
Galic
,
M.
,
Köhler
,
K.
,
Wittwer
,
F.
,
Brogiolo
,
W.
,
Hafen
,
E.
and
Stocker
,
H.
(
2008
).
Imp-L2, a putative homolog of vertebrate IGF-binding protein 7, counteracts insulin signaling in Drosophila and is essential for starvation resistance
.
J. Biol.
7
,
10
.
Honjoh
,
S.
,
Ihara
,
A.
,
Kajiwara
,
Y.
,
Yamamoto
,
T.
and
Nishida
,
E.
(
2017
).
The sexual dimorphism of dietary restriction responsiveness in Caenorhabditis elegans
.
Cell Rep.
21
,
3646
-
3652
.
Horie
,
I.
,
Abiru
,
N.
,
Eto
,
M.
,
Sako
,
A.
,
Akeshima
,
J.
,
Nakao
,
T.
,
Nakashima
,
Y.
,
Niri
,
T.
,
Ito
,
A.
,
Nozaki
,
A.
et al. 
(
2018
).
Sex differences in insulin and glucagon responses for glucose homeostasis in young healthy Japanese adults
.
J. Diabetes Invest.
9
,
1283
-
1287
.
Hubbard
,
E. J.
and
Greenstein
,
D.
(
2000
).
The Caenorhabditis elegans gonad: a test tube for cell and developmental biology
.
Dev. Dyn.
218
,
2
-
22
.
Hudry
,
B.
,
Khadayate
,
S.
and
Miguel-Aliaga
,
I.
(
2016
).
The sexual identity of adult intestinal stem cells controls organ size and plasticity
.
Nature
530
,
344
-
348
.
Jazin
,
E.
and
Cahill
,
L.
(
2010
).
Sex differences in molecular neuroscience: from fruit flies to humans
.
Nat. Rev. Neurosci.
11
,
9
-
17
.
Jiang
,
H.
and
Edgar
,
B. A.
(
2011
).
Intestinal stem cells in the adult Drosophila midgut
.
Exp. Cell Res.
317
,
2780
-
2788
.
Jiang
,
H.
and
Edgar
,
B. A.
(
2012
).
Intestinal stem cell function in Drosophila and mice
.
Curr. Opin. Genet. Dev.
22
,
354
-
360
.
Jiang
,
H.
,
Patel
,
P. H.
,
Kohlmaier
,
A.
,
Grenley
,
M. O.
,
McEwen
,
D. G.
and
Edgar
,
B. A.
(
2009
).
Cytokine/Jak/Stat signaling mediates regeneration and homeostasis in the Drosophila midgut
.
Cell
137
,
1343
-
1355
.
Karastergiou
,
K.
,
Smith
,
S. R.
,
Greenberg
,
A. S.
and
Fried
,
S. K.
(
2012
).
Sex differences in human adipose tissues - the biology of pear shape
.
Biol. Sex Differ.
3
,
13
.
Karp
,
N. A.
,
Mason
,
J.
,
Beaudet
,
A. L.
,
Benjamini
,
Y.
,
Bower
,
L.
,
Braun
,
R. E.
,
Brown
,
S. D. M.
,
Chesler
,
E. J.
,
Dickinson
,
M. E.
,
Flenniken
,
A. M.
et al. 
(
2017
).
Prevalence of sexual dimorphism in mammalian phenotypic traits
.
Nat. Commun.
8
,
15475
.
Kay
,
J. N.
,
Hannigan
,
P.
and
Kelley
,
D. B.
(
1999
).
Trophic effects of androgen: development and hormonal regulation of neuron number in a sexually dimorphic vocal motor nucleus
.
J. Neurobiol.
40
,
375
-
385
.
Khramtsova
,
E. A.
,
Davis
,
L. K.
and
Stranger
,
B. E.
(
2019
).
The role of sex in the genomics of human complex traits
.
Nat. Rev. Genet.
20
,
173
-
190
.
Kijimoto
,
T.
,
Andrews
,
J.
and
Moczek
,
A. P.
(
2010
).
Programed cell death shapes the expression of horns within and between species of horned beetles
.
Evol. Dev.
12
,
449
-
458
.
Kimura
,
K.-I.
,
Ote
,
M.
,
Tazawa
,
T.
and
Yamamoto
,
D.
(
2005
).
Fruitless specifies sexually dimorphic neural circuitry in the Drosophila brain
.
Nature
438
,
229
-
233
.
Kimura
,
K.-I.
,
Hachiya
,
T.
,
Koganezawa
,
M.
,
Tazawa
,
T.
and
Yamamoto
,
D.
(
2008
).
Fruitless and doublesex coordinate to generate male-specific neurons that can initiate courtship
.
Neuron
59
,
759
-
769
.
Kirn
,
J. R.
and
DeVoogd
,
T. J.
(
1989
).
Genesis and death of vocal control neurons during sexual differentiation in the zebra finch
.
J. Neurosci.
9
,
3176
-
3187
.
Klein
,
S. L.
and
Flanagan
,
K. L.
(
2016
).
Sex differences in immune responses
.
Nat. Rev. Immunol.
16
,
626
-
638
.
Kopp
,
A.
,
Duncan
,
I.
,
Godt
,
D.
and
Carroll
,
S. B.
(
2000
).
Genetic control and evolution of sexually dimorphic characters in Drosophila
.
Nature
408
,
553
-
559
.
Lawlor
,
D. A.
,
Ebrahim
,
S.
and
Davey Smith
,
G.
(
2001
).
Sex matters: secular and geographical trends in sex differences in coronary heart disease mortality
.
BMJ
323
,
541
-
545
.
Lawson
,
H. N.
,
Wexler
,
L. R.
,
Wnuk
,
H. K.
and
Portman
,
D. S.
(
2020
).
Dynamic, non-binary specification of sexual state in the C. elegans nervous system
.
Curr. Biol.
30
,
3617
-
3623.e3
.
Legato
,
M. J.
and
Legha
,
J. K.
(
2004
).
Gender and the heart: sex-specific differences in normal myocardial anatomy and physiology and in the experiences of some diseases of the cardiovascular system
. In
Principles of Gender-Specific Medicine (ed. M. Legato)
, pp.
185
-
192
.
Elsevier
.
Lemaitre
,
B.
and
Miguel-Aliaga
,
I.
(
2013
).
The digestive tract of Drosophila melanogaster
.
Annu. Rev. Genet.
47
,
377
-
404
.
Leopold
,
P.
and
Perrimon
,
N.
(
2007
).
Drosophila and the genetics of the internal milieu
.
Nature
450
,
186
-
188
.
Liu
,
J.-P.
,
Baker
,
J.
,
Perkins
,
A. S.
,
Robertson
,
E. J.
and
Efstratiadis
,
A.
(
1993
).
Mice carrying null mutations of the genes encoding insulin-like growth factor I (Igf-1) and type 1 IGF receptor (Igf1r)
.
Cell
75
,
59
-
72
.
Liu
,
C.-F.
,
Bingham
,
N.
,
Parker
,
K.
and
Yao
,
H. H.-C.
(
2009
).
Sex-specific roles of β-catenin in mouse gonadal development
.
Hum. Mol. Genet.
18
,
405
-
417
.
Lochab
,
A. K.
and
Extavour
,
C. G.
(
2017
).
Bone Morphogenetic Protein (BMP) signaling in animal reproductive system development and function
.
Dev. Biol.
427
,
258
-
269
.
LoMauro
,
A.
and
Aliverti
,
A.
(
2018
).
Sex differences in respiratory function
.
Breathe
14
,
131
-
140
.
Lopes-Ramos
,
C. M.
,
Chen
,
C.-Y.
,
Kuijjer
,
M. L.
,
Paulson
,
J. N.
,
Sonawane
,
A. R.
,
Fagny
,
M.
,
Platig
,
J.
,
Glass
,
K.
,
Quackenbush
,
J.
and
DeMeo
,
D. L.
(
2020
).
Sex differences in gene expression and regulatory networks across 29 human tissues
.
Cell Rep.
31
,
107795
.
Lu
,
A. T.-H.
and
Cantor
,
R. M.
(
2012
).
Allowing for sex differences increases power in a GWAS of multiplex Autism families
.
Mol. Psychiatry
17
,
215
-
222
.
Magwere
,
T.
,
Chapman
,
T.
and
Partridge
,
L.
(
2004
).
Sex differences in the effect of dietary restriction on life span and mortality rates in female and male Drosophila melanogaster
.
J. Gerontol. A Biol. Sci. Med. Sci.
59
,
B3
-
B9
.
Mahmoud
,
R.
,
Wainwright
,
S. R.
and
Galea
,
L. A. M.
(
2016
).
Sex hormones and adult hippocampal neurogenesis: regulation, implications, and potential mechanisms
.
Front. Neuroendocrinol.
41
,
129
-
152
.
Mandy
,
W.
,
Chilvers
,
R.
,
Chowdhury
,
U.
,
Salter
,
G.
,
Seigal
,
A.
and
Skuse
,
D.
(
2012
).
Sex differences in autism spectrum disorder: evidence from a large sample of children and adolescents
.
J. Autism Dev. Disord.
42
,
1304
-
1313
.
Mank
,
J. E.
(
2009
).
Sex chromosomes and the evolution of sexual dimorphism: lessons from the genome
.
Am. Nat.
173
,
141
-
150
.
Mank
,
J. E.
(
2013
).
Sex chromosome dosage compensation: definitely not for everyone
.
Trends Genet.
29
,
677
-
683
.
Mank
,
J. E.
(
2017
).
The transcriptional architecture of phenotypic dimorphism
.
Nat. Ecol. Evol.
1
,
0006
.
Mank
,
J. E.
,
Nam
,
K.
,
Brunström
,
B.
and
Ellegren
,
H.
(
2010
).
Ontogenetic complexity of sexual dimorphism and sex-specific selection
.
Mol. Biol. Evol.
27
,
1570
-
1578
.
Mao
,
K.
,
Quipildor
,
G. F.
,
Tabrizian
,
T.
,
Novaj
,
A.
,
Guan
,
F.
,
Walters
,
R. O.
,
Delahaye
,
F.
,
Hubbard
,
G. B.
,
Ikeno
,
Y.
,
Ejima
,
K.
et al. 
(
2018
).
Late-life targeting of the IGF-1 receptor improves healthspan and lifespan in female mice
.
Nat. Commun.
9
,
2394
.
Marchese
,
E.
,
Rodeghier
,
C.
,
Monson
,
R. S.
,
McCracken
,
B.
,
Shi
,
T.
,
Schrock
,
W.
,
Martellotto
,
J.
,
Oberholzer
,
J.
and
Danielson
,
K. K.
(
2015
).
Enumerating β-cells in whole human islets: sex differences and associations with clinical outcomes after islet transplantation
.
Diabetes Care
38
,
e176
-
e177
.
Martin
,
T. R.
,
Castile
,
R. G.
,
Fredberg
,
J. J.
,
Wohl
,
M. E.
and
Mead
,
J.
(
1987
).
Airway size is related to sex but not lung size in normal adults
.
J. Appl. Physiol.
63
,
2042
-
2047
.
Matoba
,
N.
,
Akiyama
,
M.
,
Ishigaki
,
K.
,
Kanai
,
M.
,
Takahashi
,
A.
,
Momozawa
,
Y.
,
Ikegawa
,
S.
,
Ikeda
,
M.
,
Iwata
,
N.
,
Hirata
,
M.
et al. 
(
2019
).
GWAS of smoking behaviour in 165,436 Japanese people reveals seven new loci and shared genetic architecture
.
Nat. Hum. Behav.
3
,
471
-
477
.
Matson
,
C. K.
and
Zarkower
,
D.
(
2012
).
Sex and the singular DM domain: insights into sexual regulation, evolution and plasticity
.
Nat. Rev. Genet.
13
,
163
-
174
.
Mattei
,
A. L.
,
Riccio
,
M. L.
,
Avila
,
F. W.
and
Wolfner
,
M. F.
(
2015
).
Integrated 3D view of postmating responses by the Drosophila melanogaster female reproductive tract, obtained by micro-computed tomography scanning
.
Proc. Natl. Acad. Sci. USA
112
,
8475
-
8480
.
Mayne
,
B. T.
,
Bianco-Miotto
,
T.
,
Buckberry
,
S.
,
Breen
,
J.
,
Clifton
,
V.
,
Shoubridge
,
C.
and
Roberts
,
C. T.
(
2016
).
Large scale gene expression meta-analysis reveals tissue-specific, sex-biased gene expression in humans
.
Front. Genet.
7
,
183
.
McDonald
,
J. M. C.
,
Nabili
,
P.
,
Thorsen
,
L.
,
Jeon
,
S.
and
Shingleton
,
A.
(
2020
).
Sex-specific plasticity and the nutritional geometry of insulin-signaling gene expression in Drosophila melanogaster
.
EvoDevo
12
,
6
.
Melé
,
M.
,
Ferreira
,
P. G.
,
Reverter
,
F.
,
DeLuca
,
D. S.
,
Monlong
,
J.
,
Sammeth
,
M.
,
Young
,
T. R.
,
Goldmann
,
J. M.
,
Pervouchine
,
D. D.
,
Sullivan
,
T. J.
et al. 
(
2015
).
The human transcriptome across tissues and individuals
.
Science
348
,
660
-
665
.
Micchelli
,
C. A.
and
Perrimon
,
N.
(
2006
).
Evidence that stem cells reside in the adult Drosophila midgut epithelium
.
Nature
439
,
475
-
479
.
Millington
,
J. W.
and
Rideout
,
E. J.
(
2018
).
Sex differences in Drosophila development and physiology
.
Curr. Opin. Physiol.
6
,
46
-
56
.
Millington
,
J. W.
,
Brownrigg
,
G. P.
,
Basner-Collins
,
P. J.
,
Sun
,
Z.
and
Rideout
,
E. J.
(
2021a
).
Genetic manipulation of insulin/insulin-like growth factor signaling pathway activity has sex-biased effects on Drosophila body size
.
G3
11
,
jkaa067
.
Millington
,
J. W.
,
Brownrigg
,
G. P.
,
Chao
,
C.
,
Sun
,
Z.
,
Basner-Collins
,
P. J.
,
Wat
,
L. W.
,
Hudry
,
B.
,
Miguel-Aliaga
,
I.
and
Rideout
,
E. J.
(
2021b
).
Female-biased upregulation of insulin pathway activity mediates the sex difference in Drosophila body size plasticity
.
eLife
10
,
e58341
.
Mitchell
,
S. J.
,
Madrigal-Matute
,
J.
,
Scheibye-Knudsen
,
M.
,
Fang
,
E.
,
Aon
,
M.
,
González-Reyes
,
J. A.
,
Cortassa
,
S.
,
Kaushik
,
S.
,
Gonzalez-Freire
,
M.
,
Patel
,
B.
et al. 
(
2016
).
Effects of sex, strain, and energy intake on hallmarks of aging in mice
.
Cell Metab.
23
,
1093
-
1112
.
Morrow
,
E. H.
(
2015
).
The evolution of sex differences in disease
.
Biol. Sex Differ.
6
,
5
.
Nakada
,
D.
,
Oguro
,
H.
,
Levi
,
B. P.
,
Ryan
,
N.
,
Kitano
,
A.
,
Saitoh
,
Y.
,
Takeichi
,
M.
,
Wendt
,
G. R.
and
Morrison
,
S. J.
(
2014
).
Oestrogen increases haematopoietic stem-cell self-renewal in females and during pregnancy
.
Nature
505
,
555
-
558
.
Narayan
,
A.
,
Venkatachalam
,
V.
,
Durak
,
O.
,
Reilly
,
D. K.
,
Bose
,
N.
,
Schroeder
,
F. C.
,
Samuel
,
A. D. T.
,
Srinivasan
,
J.
and
Sternberg
,
P. W.
(
2016
).
Contrasting responses within a single neuron class enable sex-specific attraction in Caenorhabditis elegans
.
Proc. Natl. Acad. Sci. USA
113
,
E1392
-
E1401
.
Nef
,
S.
and
Parada
,
L. F.
(
2000
).
Hormones in male sexual development
.
Genes Dev.
14
,
3075
-
3086
.
Niitsu
,
S.
,
Toga
,
K.
,
Tomizuka
,
S.
,
Maekawa
,
K.
,
Machida
,
R.
and
Kamito
,
T.
(
2014
).
Ecdysteroid-induced programmed cell death is essential for sex-specific wing degeneration of the wingless-female winter moth
.
PLoS ONE
9
,
e89435
.
Noguerol
,
J.
,
Roustan
,
P.-J.
,
N'Taye
,
M.
,
Delcombel
,
L.
,
Rolland
,
C.
,
Guiraud
,
L.
,
Sagnat
,
D.
,
Edir
,
A.
,
Bonnart
,
C.
,
Denadai-Souza
,
A.
et al. 
(
2019
).
Sexual dimorphism in PAR2-dependent regulation of primitive colonic cells
.
Biol. Sex Differ.
10
,
47
.
Noiret
,
A.
,
Puch
,
L.
,
Riffaud
,
C.
,
Costantini
,
D.
,
Riou
,
J.-F.
,
Aujard
,
F.
and
Terrien
,
J.
(
2020
).
Sex-specific response to caloric restriction after reproductive investment in microcebus murinus: an integrative approach
.
Front. Physiol.
11
,
506
.
Nottebohm
,
F.
and
Arnold
,
A. P.
(
1976
).
Sexual dimorphism in vocal control areas of the songbird brain
.
Science
194
,
211
-
213
.
Ober
,
C.
,
Loisel
,
D. A.
and
Gilad
,
Y.
(
2008
).
Sex-specific genetic architecture of human disease
.
Nat. Rev. Genet.
9
,
911
-
922
.
Offen
,
N.
,
Blum
,
N.
,
Meyer
,
A.
and
Begemann
,
G.
(
2008
).
Fgfr1 signalling in the development of a sexually selected trait in vertebrates, the sword of swordtail fish
.
BMC Dev. Biol.
8
,
98
.
Ogawa
,
R.
,
Mizuno
,
H.
,
Watanabe
,
A.
,
Migita
,
M.
,
Hyakusoku
,
H.
and
Shimada
,
T.
(
2004
).
Adipogenic differentiation by adipose-derived stem cells harvested from GFP transgenic mice—including relationship of sex differences
.
Biochem. Biophys. Res. Commun.
319
,
511
-
517
.
Ohlstein
,
B.
and
Spradling
,
A.
(
2006
).
The adult Drosophila posterior midgut is maintained by pluripotent stem cells
.
Nature
439
,
470
-
474
.
Oldham
,
S.
and
Hafen
,
E.
(
2003
).
Insulin/IGF and target of rapamycin signaling: a TOR de force in growth control
.
Trends Cell Biol.
13
,
79
-
85
.
Oliva
,
M.
,
Muñoz-Aguirre
,
M.
,
Kim-Hellmuth
,
S.
,
Wucher
,
V.
,
Gewirtz
,
A. D. H.
,
Cotter
,
D. J.
,
Parsana
,
P.
,
Kasela
,
S.
,
Balliu
,
B.
,
Viñuela
,
A.
et al. 
(
2020
).
The impact of sex on gene expression across human tissues
.
Science
369
,
eaba3066
.
Parchami
,
A.
and
Kusha
,
S.
(
2015
).
Effect of sex on histomorphometric properties of Langerhans islets in native chickens
.
Vet. Res. Forum
6
,
327
-
330
.
Pavlou
,
H. J.
,
Lin
,
A. C.
,
Neville
,
M. C.
,
Nojima
,
T.
,
Diao
,
F.
,
Chen
,
B. E.
,
White
,
B. H.
and
Goodwin
,
S. F.
(
2016
).
Neural circuitry coordinating male copulation
.
eLife
5
,
e20713
.
Penn
,
J. K. M.
and
Schedl
,
P.
(
2007
).
The master switch gene sex-lethal promotes female development by negatively regulating the N-signaling pathway
.
Dev. Cell
12
,
275
-
286
.
Perry
,
J. C.
,
Harrison
,
P. W.
and
Mank
,
J. E.
(
2014
).
The ontogeny and evolution of sex-biased gene expression in Drosophila melanogaster
.
Mol. Biol. Evol.
31
,
1206
-
1219
.
Pitnick
,
S.
,
Wolfner
,
M. F.
and
Suarez
,
S. S
. (
2009
).
Ejaculate–female and sperm–female interactions
. In
Sperm Biology
(ed.
S. S.
Pitnick
,
D. J.
Hosken
and
T.
Birkhead
), pp.
247
-
304
.
Elsevier
.
Poiani
,
A.
(
2006
).
Complexity of seminal fluid: a review
.
Behav. Ecol. Sociobiol.
60
,
289
-
310
.
Pomatto
,
L. C. D.
,
Carney
,
C.
,
Shen
,
B.
,
Wong
,
S.
,
Halaszynski
,
K.
,
Salomon
,
M. P.
,
Davies
,
K. J. A.
and
Tower
,
J.
(
2017
).
The mitochondrial lon protease is required for age-specific and sex-specific adaptation to oxidative stress
.
Curr. Biol.
27
,
1
-
15
.
Portman
,
D. S.
(
2007
).
Genetic control of sex differences in C. elegans neurobiology and behavior
.
Adv. Genet.
59
,
1
-
37
.
Powell
,
D. L.
,
Payne
,
C.
,
Banerjee
,
S. M.
,
Keegan
,
M.
,
Bashkirova
,
E.
,
Cui
,
R.
,
Andolfatto
,
P.
,
Rosenthal
,
G. G.
and
Schumer
,
M.
(
2021
).
The genetic architecture of variation in the sexually selected sword ornament and its evolution in hybrid populations
.
Curr. Biol.
31
,
923
-
935.e11
.
Powell-Braxton
,
L.
,
Hollingshead
,
P.
,
Warburton
,
C.
,
Dowd
,
M.
,
Pitts-Meek
,
S.
,
Dalton
,
D.
,
Gillett
,
N.
and
Stewart
,
T. A.
(
1993
).
IGF-I is required for normal embryonic growth in mice
.
Genes Dev.
7
,
2609
-
2617
.
Randall
,
J. C.
,
Winkler
,
T. W.
,
Kutalik
,
Z.
,
Berndt
,
S. I.
,
Jackson
,
A. U.
,
Monda
,
K. L.
,
Kilpeläinen
,
T. O.
,
Esko
,
T.
,
Mägi
,
R.
,
Li
,
S.
et al. 
(
2013
).
Sex-stratified genome-wide association studies including 270,000 individuals show sexual dimorphism in genetic loci for anthropometric traits
.
PLoS Genet.
9
,
e1003500
.
Regan
,
J. C.
,
Khericha
,
M.
,
Dobson
,
A. J.
,
Bolukbasi
,
E.
,
Rattanavirotkul
,
N.
and
Partridge
,
L.
(
2016
).
Sex difference in pathology of the ageing gut mediates the greater response of female lifespan to dietary restriction
.
eLife
5
,
e10956
.
Reiff
,
T.
,
Jacobson
,
J.
,
Cognigni
,
P.
,
Antonello
,
Z.
,
Ballesta
,
E.
,
Tan
,
K. J.
,
Yew
,
J. Y.
,
Dominguez
,
M.
and
Miguel-Aliaga
,
I.
(
2015
).
Endocrine remodelling of the adult intestine sustains reproduction in Drosophila
.
eLife
4
,
e06930
.
Rezania
,
A.
,
Bruin
,
J. E.
,
Riedel
,
M. J.
,
Mojibian
,
M.
,
Asadi
,
A.
,
Xu
,
J.
,
Gauvin
,
R.
,
Narayan
,
K.
,
Karanu
,
F.
,
O'Neil
,
J. J.
et al. 
(
2012
).
Maturation of human embryonic stem cell-derived pancreatic progenitors into functional islets capable of treating pre-existing diabetes in mice
.
Diabetes
61
,
2016
-
2029
.
Rezával
,
C.
,
Nojima
,
T.
,
Neville
,
M. C.
,
Lin
,
A. C.
and
Goodwin
,
S. F.
(
2014
).
Sexually dimorphic octopaminergic neurons modulate female postmating behaviors in Drosophila
.
Curr. Biol.
24
,
725
-
730
.
Ribeiro
,
C.
and
Dickson
,
B. J.
(
2010
).
Sex peptide receptor and neuronal TOR/S6K signaling modulate nutrient balancing in Drosophila
.
Curr. Biol.
20
,
1000
-
1005
.
Richardson
,
N. E.
,
Konon
,
E. N.
,
Schuster
,
H. S.
,
Mitchell
,
A. T.
,
Boyle
,
C.
,
Rodgers
,
A. C.
,
Finke
,
M.
,
Haider
,
L. R.
,
Yu
,
D.
,
Flores
,
V.
et al. 
(
2021
).
Lifelong restriction of dietary branched-chain amino acids has sex-specific benefits for frailty and life span in mice
.
Nat. Aging
1
,
73
-
86
.
Rideout
,
E. J.
,
Billeter
,
J.-C.
and
Goodwin
,
S. F.
(
2007
).
The sex-determination genes fruitless and doublesex specify a neural substrate required for courtship song
.
Curr. Biol.
17
,
1473
-
1478
.
Rideout
,
E. J.
,
Dornan
,
A. J.
,
Neville
,
M. C.
,
Eadie
,
S.
and
Goodwin
,
S. F.
(
2010
).
Control of sexual differentiation and behavior by the doublesex gene in Drosophila melanogaster
.
Nat. Neurosci.
13
,
458
-
466
.
Rideout
,
E. J.
,
Narsaiya
,
M. S.
and
Grewal
,
S. S.
(
2015
).
The sex determination gene transformer regulates male-female differences in Drosophila body size
.
PLoS Genet.
11
,
e1005683
.
Robinett
,
C. C.
,
Vaughan
,
A. G.
,
Knapp
,
J.-M.
and
Baker
,
B. S.
(
2010
).
Sex and the single cell. II. There is a time and place for sex
.
PLoS Biol.
8
,
e1000365
.
Rodríguez-Martínez
,
H.
,
Kvist
,
U.
,
Ernerudh
,
J.
,
Sanz
,
L.
and
Calvete
,
J. J.
(
2011
).
Seminal plasma proteins: what role do they play?
Am. J. Reprod. Immunol.
66
Suppl. 1
,
11
-
22
.
Ross
,
J. M.
,
Kalis
,
A. K.
,
Murphy
,
M. W.
and
Zarkower
,
D.
(
2005
).
The DM domain protein MAB-3 promotes sex-specific neurogenesis in C. elegans by regulating bHLH proteins
.
Dev. Cell
8
,
881
-
892
.
Rubinstein
,
C. D.
and
Wolfner
,
M. F.
(
2013
).
Drosophila seminal protein ovulin mediates ovulation through female octopamine neuronal signaling
.
Proc. Natl. Acad. Sci. USA
110
,
17420
-
17425
.
Ryan
,
D. A.
,
Miller
,
R. M.
,
Lee
,
K.
,
Neal
,
S. J.
,
Fagan
,
K. A.
,
Sengupta
,
P.
and
Portman
,
D. S.
(
2014
).
Sex, age, and hunger regulate behavioral prioritization through dynamic modulation of chemoreceptor expression
.
Curr. Biol.
24
,
2509
-
2517
.
Saad
,
M. F.
,
Damani
,
S.
,
Gingerich
,
R. L.
,
Riad-Gabriel
,
M. G.
,
Khan
,
A.
,
Boyadjian
,
R.
,
Jinagouda
,
S. D.
,
el-Tawil
,
K.
,
Rude
,
R. K.
and
Kamdar
,
V.
(
1997
).
Sexual dimorphism in plasma leptin concentration
.
J. Clin. Endocrinol. Metab.
82
,
579
-
584
.
Saber
,
N.
,
Bruin
,
J. E.
,
O'Dwyer
,
S.
,
Schuster
,
H.
,
Rezania
,
A.
and
Kieffer
,
T. J.
(
2018
).
Sex differences in maturation of human embryonic stem cell-derived β cells in mice
.
Endocrinology
159
,
1827
-
1841
.
Sakai
,
N.
,
Iwata
,
R.
,
Yokoi
,
S.
,
Butcher
,
R. A.
,
Clardy
,
J.
,
Tomioka
,
M.
and
Iino
,
Y.
(
2013
).
A sexually conditioned switch of chemosensory behavior in C. elegans
.
PLoS ONE
8
,
e68676
.
Sammut
,
M.
,
Cook
,
S. J.
,
Nguyen
,
K. C. Q.
,
Felton
,
T.
,
Hall
,
D. H.
,
Emmons
,
S. W.
,
Poole
,
R. J.
and
Barrios
,
A.
(
2015
).
Glia-derived neurons are required for sex-specific learning in C. elegans
.
Nature
526
,
385
-
390
.
Sanchez
,
L.
,
Gorfinkiel
,
N.
and
Guerrero
,
I.
(
2001
).
Sex determination genes control the development of the Drosophila genital disc, modulating the response to Hedgehog, Wingless and Decapentaplegic signals
.
Development
128
,
1033
-
1043
.
Sanders
,
L. E.
and
Arbeitman
,
M. N.
(
2008
).
Doublesex establishes sexual dimorphism in the Drosophila central nervous system in an isoform-dependent manner by directing cell number
.
Dev. Biol.
320
,
378
-
390
.
Sandkam
,
B. A.
,
Almeida
,
P.
,
Darolti
,
I.
,
Furman
,
B. L. S.
,
van der Bijl
,
W.
,
Morris
,
J.
,
Bourne
,
G. R.
,
Breden
,
F.
and
Mank
,
J. E.
(
2021
).
Extreme Y chromosome polymorphism corresponds to five male reproductive morphs of a freshwater fish
.
Nat. Ecol. Evol.
5
,
939
-
948
.
Schartl
,
M.
,
Kneitz
,
S.
,
Ormanns
,
J.
,
Schmidt
,
C.
,
Anderson
,
J. L.
,
Amores
,
A.
,
Catchen
,
J.
,
Wilson
,
C.
,
Geiger
,
D.
,
Du
,
K.
et al. 
(
2021
).
The developmental and genetic architecture of the sexually selected male ornament of swordtails
.
Curr. Biol.
31
,
911
-
922.e4
.
Sekido
,
R.
and
Lovell-Badge
,
R.
(
2013
).
Genetic control of testis development
.
Sex. Dev.
7
,
21
-
32
.
Shah
,
T.
and
Rogers
,
M.
(
2018
).
Unanswered questions regarding sex and BMP/TGF-β signaling
.
J. Dev. Biol.
6
,
14
.
Shingleton
,
A. W.
,
Masandika
,
J. R.
,
Thorsen
,
L. S.
,
Zhu
,
Y.
and
Mirth
,
C. K.
(
2017
).
The sex-specific effects of diet quality versus quantity on morphology in Drosophila melanogaster
.
R. Soc. Open Sci.
4
,
170375
.
Shirangi
,
T. R.
,
Wong
,
A. M.
,
Truman
,
J. W.
and
Stern
,
D. L.
(
2016
).
Doublesex regulates the connectivity of a neural circuit controlling Drosophila male courtship song
.
Dev. Cell
37
,
533
-
544
.
Shungin
,
D.
,
Winkler
,
T. W.
,
Croteau-Chonka
,
D. C.
,
Ferreira
,
T.
,
Locke
,
A. E.
,
Mägi
,
R.
,
Strawbridge
,
R. J.
,
Pers
,
T. H.
,
Fischer
,
K.
,
Justice
,
A. E.
et al. 
(
2015
).
New genetic loci link adipose and insulin biology to body fat distribution
.
Nature
518
,
187
-
196
.
Sieber
,
M. H.
and
Spradling
,
A. C.
(
2015
).
Steroid signaling establishes a female metabolic state and regulates SREBP to control oocyte lipid accumulation
.
Curr. Biol.
25
,
993
-
1004
.
Soldin
,
O. P.
and
Mattison
,
D. R.
(
2009
).
Sex differences in pharmacokinetics and pharmacodynamics
.
Clin. Pharmacokinet.
48
,
143
-
157
.
Stokes
,
D. R.
,
Malamud
,
J. G.
and
Schreihofer
,
D. A.
(
1994
).
Gender specific developmental transformation of a cockroach bifunctional muscle
.
J. Exp. Zool.
268
,
364
-
376
.
Sulston
,
J. E.
,
Schierenberg
,
E.
,
White
,
J. G.
and
Thomson
,
J. N.
(
1983
).
The embryonic cell lineage of the nematode Caenorhabditis elegans
.
Dev. Biol.
100
,
64
-
119
.
Swain
,
A.
and
Lovell-Badge
,
R.
(
1999
).
Mammalian sex determination: a molecular drama
.
Genes Dev.
13
,
755
-
767
.
Tatar
,
M.
,
Kopelman
,
A.
,
Epstein
,
D.
,
Tu
,
M.-P.
,
Yin
,
C.-M.
and
Garofalo
,
R. S.
(
2001
).
A mutant Drosophila insulin receptor homolog that extends life-span and impairs neuroendocrine function
.
Science
292
,
107
-
110
.
Taylor
,
B. J.
and
Truman
,
J. W.
(
1992
).
Commitment of abdominal neuroblasts in Drosophila to a male or female fate is dependent on genes of the sex-determining hierarchy
.
Development
114
,
625
-
642
.
Taylor
,
R. W.
,
Grant
,
A. M.
,
Williams
,
S. M.
and
Goulding
,
A.
(
2010
).
Sex differences in regional body fat distribution from pre- to postpuberty
.
Obesity
18
,
1410
-
1416
.
Teleman
,
A. A.
(
2010
).
Molecular mechanisms of metabolic regulation by insulin in Drosophila
.
Biochem. J.
425
,
13
-
26
.
Templeman
,
N. M.
,
Flibotte
,
S.
,
Chik
,
J. H. L.
,
Sinha
,
S.
,
Lim
,
G. E.
,
Foster
,
L. J.
,
Nislow
,
C.
and
Johnson
,
J. D.
(
2017
).
Reduced circulating insulin enhances insulin sensitivity in old mice and extends lifespan
.
Cell Rep.
20
,
451
-
463
.
Testa
,
N. D.
and
Dworkin
,
I.
(
2016
).
The sex-limited effects of mutations in the EGFR and TGF-β signaling pathways on shape and size sexual dimorphism and allometry in the Drosophila wing
.
Dev. Genes Evol.
226
,
159
-
171
.
Testa
,
N. D.
,
Ghosh
,
S. M.
and
Shingleton
,
A. W.
(
2013
).
Sex-specific weight loss mediates sexual size dimorphism in Drosophila melanogaster
.
PLoS ONE
8
,
e58936
.
Towbin
,
J. A.
,
Hejtmancik
,
J. F.
,
Brink
,
P.
,
Gelb
,
B.
,
Zhu
,
X. M.
,
Chamberlain
,
J. S.
,
McCabe
,
E. R.
and
Swift
,
M.
(
1993
).
X-linked dilated cardiomyopathy. Molecular genetic evidence of linkage to the Duchenne muscular dystrophy (dystrophin) gene at the Xp21 locus
.
Circulation
87
,
1854
-
1865
.
Tower
,
J.
,
Pomatto
,
L. C. D.
and
Davies
,
K. J. A.
(
2020
).
Sex differences in the response to oxidative and proteolytic stress
.
Redox Biol.
31
,
101488
.
Traglia
,
M.
,
Bseiso
,
D.
,
Gusev
,
A.
,
Adviento
,
B.
,
Park
,
D. S.
,
Mefford
,
J. A.
,
Zaitlen
,
N.
and
Weiss
,
L. A.
(
2017
).
Genetic mechanisms leading to sex differences across common diseases and anthropometric traits
.
Genetics
205
,
979
-
992
.
Tükel
,
R.
,
Ertekin
,
E.
,
Batmaz
,
S.
,
Alyanak
,
F.
,
Sözen
,
A.
,
Aslantaş
,
B.
,
Atli
,
H.
and
Ozyildirim
,
I.
(
2005
).
Influence of age of onset on clinical features in obsessive-compulsive disorder
.
Depress. Anxiety
21
,
112
-
117
.
Tweedie
,
S.
,
Ashburner
,
M.
,
Falls
,
K.
,
Leyland
,
P.
,
McQuilton
,
P.
,
Marygold
,
S.
,
Millburn
,
G.
,
Osumi-Sutherland
,
D.
,
Schroeder
,
A.
,
Seal
,
R.
et al. 
(
2009
).
FlyBase: enhancing Drosophila gene ontology annotations
.
Nucleic Acids Res.
37
,
D555
-
D559
.
van der Bijl
,
W.
and
Mank
,
J. E.
(
2021
).
Widespread cryptic variation in genetic architecture between the sexes
.
Evol. Lett.
5
,
359
-
369
.
van der Kroon
,
I.
,
Woliner-van der Weg
,
W.
,
Brom
,
M.
,
Joosten
,
L.
,
Frielink
,
C.
,
Konijnenberg
,
M. W.
,
Visser
,
E. P.
and
and Gotthardt
,
M.
(
2017
).
Whole organ and islet of Langerhans dosimetry for calculation of absorbed doses resulting from imaging with radiolabeled exendin
.
Sci. Rep.
7
,
39800
.
Vargas
,
M. A.
,
Luo
,
N.
,
Yamaguchi
,
A.
and
Kapahi
,
P.
(
2010
).
A role for S6 kinase and serotonin in postmating dietary switch and balance of nutrients in D. melanogaster
.
Curr. Biol.
20
,
1006
-
1011
.
Walker
,
S. J.
,
Corrales-Carvajal
,
V. M.
and
Ribeiro
,
C.
(
2015
).
Postmating circuitry modulates salt taste processing to increase reproductive output in Drosophila
.
Curr. Biol.
25
,
2621
-
2630
.
Wang
,
C.
,
He
,
Y.
,
Xu
,
P.
,
Yang
,
Y.
,
Saito
,
K.
,
Xia
,
Y.
,
Yan
,
X.
,
Hinton
,
A.
, Jr.
,
Yan
,
C.
,
Ding
,
H.
et al. 
(
2018
).
TAp63 contributes to sexual dimorphism in POMC neuron functions and energy homeostasis
.
Nat. Commun.
9
,
1544
.
Wang
,
M.
,
Gorelick
,
F.
and
Bhargava
,
A.
(
2021
).
Sex differences in the exocrine pancreas and associated diseases
.
Cell. Mol. Gastroenterol. Hepatol.
12
,
427
-
441
.
Wat
,
L. W.
,
Chao
,
C.
,
Bartlett
,
R.
,
Buchanan
,
J. L.
,
Millington
,
J. W.
,
Chih
,
H. J.
,
Chowdhury
,
Z. S.
,
Biswas
,
P.
,
Huang
,
V.
,
Shin
,
L. J.
et al. 
(
2020
).
A role for triglyceride lipase brummer in the regulation of sex differences in Drosophila fat storage and breakdown
.
PLoS Biol.
18
,
e3000595
.
Wawersik
,
M.
,
Milutinovich
,
A.
,
Casper
,
A. L.
,
Matunis
,
E.
,
Williams
,
B.
and
Van Doren
,
M.
(
2005
).
Somatic control of germline sexual development is mediated by the JAK/STAT pathway
.
Nature
436
,
563
-
567
.
Whitworth
,
C.
,
Jimenez
,
E.
and
Van Doren
,
M.
(
2012
).
Development of sexual dimorphism in the Drosophila testis
.
Spermatogenesis
2
,
129
-
136
.
Williams
,
T. M.
and
Carroll
,
S. B.
(
2009
).
Genetic and molecular insights into the development and evolution of sexual dimorphism
.
Nat. Rev. Genet.
10
,
797
-
804
.
Windley
,
S. P.
and
Wilhelm
,
D.
(
2016
).
Signaling pathways involved in mammalian sex determination and gonad development
.
Sex. Dev.
9
,
297
-
315
.
Winkler
,
T. W.
,
Justice
,
A. E.
,
Graff
,
M.
,
Barata
,
L.
,
Feitosa
,
M. F.
,
Chu
,
S.
,
Czajkowski
,
J.
,
Esko
,
T.
,
Fall
,
T.
,
Kilpeläinen
,
T. O.
et al. 
(
2015
).
The influence of age and sex on genetic associations with adult body size and shape: a large-scale genome-wide interaction study
.
PLoS Genet.
11
,
e1005378
.
Wolfner
,
M. F.
(
1997
).
Tokens of love: functions and regulation of Drosophila male accessory gland products
.
Insect Biochem. Mol. Biol.
27
,
179
-
192
.
Woodling
,
N. S.
,
Aleyakpo
,
B.
,
Dyson
,
M. C.
,
Minkley
,
L. J.
,
Rajasingam
,
A.
,
Dobson
,
A. J.
,
Leung
,
K. H. C.
,
Pomposova
,
S.
,
Fuentealba
,
M.
,
Alic
,
N.
et al. 
(
2020
).
The neuronal receptor tyrosine kinase Alk is a target for longevity
.
Aging Cell
19
,
e13137
.
Xie
,
J. Z.
,
Tarczy-Hornoch
,
K.
,
Lin
,
J.
,
Cotter
,
S. A.
,
Torres
,
M.
and
Varma
,
R.
and for the Multi-Ethnic Pediatric Eye Disease Study Group
. (
2014
).
Color vision deficiency in preschool children: the multi-ethnic pediatric eye disease study
.
Ophthalmology
121
,
1469
-
1474
.
Xu
,
J.
,
Gontier
,
G.
,
Chaker
,
Z.
,
Lacube
,
P.
,
Dupont
,
J.
and
Holzenberger
,
M.
(
2014
).
Longevity effect of IGF-1R+/− mutation depends on genetic background-specific receptor activation
.
Aging Cell
13
,
19
-
28
.
Yu
,
H.
,
Mistry
,
J.
,
Nicar
,
M. J.
,
Khosravi
,
M. J.
,
Diamandis
,
A.
,
van Doorn
,
J.
and
Juul
,
A.
(
1999
).
Insulin-like growth factors (IGF-I, free IGF-I and IGF-II) and insulin-like growth factor binding proteins (IGFBP-2, IGFBP-3, IGFBP-6, and ALS) in blood circulation
.
J. Clin. Lab. Anal.
13
,
166
-
172
.
Zajitschek
,
S. R. K.
,
Zajitschek
,
F.
,
Bonduriansky
,
R.
,
Brooks
,
R. C.
,
Cornwell
,
W.
,
Falster
,
D. S.
,
Lagisz
,
M.
,
Mason
,
J.
,
Senior
,
A. M.
,
Noble
,
D. W. A.
et al. 
(
2020
).
Sexual dimorphism in trait variability and its eco-evolutionary and statistical implications
.
eLife
9
,
e63170
.
Zarkower
,
D.
(
2001
).
Establishing sexual dimorphism: conservation amidst diversity?
Nat. Rev. Genet.
2
,
175
-
185
.
Zarkower
,
D.
(
2013
).
DMRT genes in vertebrate gametogenesis
.
Curr. Top. Dev. Biol.
102
,
327
-
356
.
Zhang
,
J.-M.
,
Konkle
,
A. T. M.
,
Zup
,
S. L.
and
McCarthy
,
M. M.
(
2008
).
Impact of sex and hormones on new cells in the developing rat hippocampus: a novel source of sex dimorphism?
Eur. J. Neurosci.
27
,
791
-
800
.
Zhou
,
W.
,
Davis
,
E. A.
,
Li
,
K.
,
Nowak
,
R. A.
and
Dailey
,
M. J.
(
2018
).
Sex differences influence intestinal epithelial stem cell proliferation independent of obesity
.
Physiol. Rep.
6
,
e13746
.
Zipper
,
L.
,
Jassmann
,
D.
,
Burgmer
,
S.
,
Görlich
,
B.
and
Reiff
,
T.
(
2020
).
Ecdysone steroid hormone remote controls intestinal stem cell fate decisions via the PPARγ-homolog Eip75B in Drosophila
.
eLife
9
,
e55795
.

Competing interests

The authors declare no competing or financial interests.