Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
bioRxiv ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38915619

ABSTRACT

Animals need to integrate information across neuronal networks that direct reproductive behaviors and circadian rhythms. In Drosophila, the master regulatory transcription factors that direct courtship behaviors and circadian rhythms are co-expressed in a small set of neurons. In this study we investigate the role of these neurons in both males and females. We find sex-differences in the number of these fruitless and Clock -expressing neurons ( fru ∩ Clk neurons) that is regulated by male-specific Fru. We assign the fru ∩ Clk neurons to the electron microscopy connectome that provides high resolution structural information. We also discover sex-differences in the number of fru -expressing neurons that are post-synaptic targets of Clk -expressing neurons, with more post-synaptic targets in males. When fru ∩ Clk neurons are activated or silenced, males have a shorter period length. Activation of fru ∩ Clk neurons also changes the rate a courtship behavior is performed. We find that activation and silencing fru ∩ Clk neurons impacts the molecular clock in the sLNv master pacemaker neurons, in a cell-nonautonomous manner. These results reveal how neurons that subserve the two processes, reproduction and circadian rhythms, can impact behavioral outcomes in a sex-specific manner.

2.
Elife ; 122023 02 01.
Article in English | MEDLINE | ID: mdl-36724009

ABSTRACT

Drosophila melanogaster reproductive behaviors are orchestrated by fruitless neurons. We performed single-cell RNA-sequencing on pupal neurons that produce sex-specifically spliced fru transcripts, the fru P1-expressing neurons. Uniform Manifold Approximation and Projection (UMAP) with clustering generates an atlas containing 113 clusters. While the male and female neurons overlap in UMAP space, more than half the clusters have sex differences in neuron number, and nearly all clusters display sex-differential expression. Based on an examination of enriched marker genes, we annotate clusters as circadian clock neurons, mushroom body Kenyon cell neurons, neurotransmitter- and/or neuropeptide-producing, and those that express doublesex. Marker gene analyses also show that genes that encode members of the immunoglobulin superfamily of cell adhesion molecules, transcription factors, neuropeptides, neuropeptide receptors, and Wnts have unique patterns of enriched expression across the clusters. In vivo spatial gene expression links to the clusters are examined. A functional analysis of fru P1 circadian neurons shows they have dimorphic roles in activity and period length. Given that most clusters are comprised of male and female neurons indicates that the sexes have fru P1 neurons with common gene expression programs. Sex-specific expression is overlaid on this program, to build the potential for vastly different sex-specific behaviors.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Female , Male , Drosophila melanogaster/physiology , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Transcriptome , Sexual Behavior, Animal/physiology , Neurons/physiology , Sex Characteristics , Nerve Tissue Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Science ; 375(6584): eabk2432, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35239393

ABSTRACT

For more than 100 years, the fruit fly Drosophila melanogaster has been one of the most studied model organisms. Here, we present a single-cell atlas of the adult fly, Tabula Drosophilae, that includes 580,000 nuclei from 15 individually dissected sexed tissues as well as the entire head and body, annotated to >250 distinct cell types. We provide an in-depth analysis of cell type-related gene signatures and transcription factor markers, as well as sexual dimorphism, across the whole animal. Analysis of common cell types between tissues, such as blood and muscle cells, reveals rare cell types and tissue-specific subtypes. This atlas provides a valuable resource for the Drosophila community and serves as a reference to study genetic perturbations and disease models at single-cell resolution.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Transcriptome , Animals , Cell Nucleus/metabolism , Databases, Genetic , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Female , Gene Expression Regulation , Gene Regulatory Networks , Genes, Insect , Male , RNA-Seq , Sex Characteristics , Single-Cell Analysis , Transcription Factors/genetics
4.
BMC Res Notes ; 14(1): 236, 2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34162407

ABSTRACT

OBJECTIVES: Arachnids have fascinating and unique biology, particularly for questions on sex differences and behavior, creating the potential for development of powerful emerging models in this group. Recent advances in genomic techniques have paved the way for a significant increase in the breadth of genomic studies in non-model organisms. One growing area of research is comparative transcriptomics. When phylogenetic relationships to model organisms are known, comparative genomic studies provide context for analysis of homologous genes and pathways. The goal of this study was to lay the groundwork for comparative transcriptomics of sex differences in the brain of wolf spiders, a non-model organism of the pyhlum Euarthropoda, by generating transcriptomes and analyzing gene expression. DATA DESCRIPTION: To examine sex-differential gene expression, short read transcript sequencing and de novo transcriptome assembly were performed. Messenger RNA was isolated from brain tissue of male and female subadult and mature wolf spiders (Schizocosa ocreata). The raw data consist of sequences for the two different life stages in each sex. Computational analyses on these data include de novo transcriptome assembly and differential expression analyses. Sample-specific and combined transcriptomes, gene annotations, and differential expression results are described in this data note and are available from publicly-available databases.


Subject(s)
Spiders , Transcriptome , Animals , Brain , Female , Gene Expression Profiling , Male , Molecular Sequence Annotation , Phylogeny , Spiders/genetics , Transcriptome/genetics
5.
PLoS Genet ; 17(4): e1009240, 2021 04.
Article in English | MEDLINE | ID: mdl-33901168

ABSTRACT

Examining the role of chromatin modifications and gene expression in neurons is critical for understanding how the potential for behaviors are established and maintained. We investigate this question by examining Drosophila melanogaster fru P1 neurons that underlie reproductive behaviors in both sexes. We developed a method to purify cell-type-specific chromatin (Chromatag), using a tagged histone H2B variant that is expressed using the versatile Gal4/UAS gene expression system. Here, we use Chromatag to evaluate five chromatin modifications, at three life stages in both sexes. We find substantial changes in chromatin modification profiles across development and fewer differences between males and females. Additionally, we find chromatin modifications that persist in different sets of genes from pupal to adult stages, which may point to genes important for cell fate determination in fru P1 neurons. We generated cell-type-specific RNA-seq data sets, using translating ribosome affinity purification (TRAP). We identify actively translated genes in fru P1 neurons, revealing novel stage- and sex-differences in gene expression. We also find chromatin modification enrichment patterns that are associated with gene expression. Next, we use the chromatin modification data to identify cell-type-specific super-enhancer-containing genes. We show that genes with super-enhancers in fru P1 neurons differ across development and between the sexes. We validated that a set of genes are expressed in fru P1 neurons, which were chosen based on having a super-enhancer and TRAP-enriched expression in fru P1 neurons.


Subject(s)
Chromatin/genetics , Drosophila Proteins/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Sexual Behavior, Animal/physiology , Transcription Factors/genetics , Animals , Cell Lineage/genetics , Chromatin Assembly and Disassembly/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental/genetics , Pupa/genetics , Pupa/growth & development , RNA-Seq
6.
Elife ; 102021 02 22.
Article in English | MEDLINE | ID: mdl-33616528

ABSTRACT

Drosophila reproductive behaviors are directed by fruitless neurons. A reanalysis of genomic studies shows that genes encoding dpr and DIP immunoglobulin superfamily (IgSF) members are expressed in fru P1 neurons. We find that each fru P1 and dpr/DIP (fru P1 ∩ dpr/DIP) overlapping expression pattern is similar in both sexes, but there are dimorphisms in neuronal morphology and cell number. Behavioral studies of fru P1 ∩ dpr/DIP perturbation genotypes indicate that the mushroom body functions together with the lateral protocerebral complex to direct courtship behavior. A single-cell RNA-seq analysis of fru P1 neurons shows that many DIPs have high expression in a small set of neurons, whereas the dprs are often expressed in a larger set of neurons at intermediate levels, with a myriad of dpr/DIP expression combinations. Functionally, we find that perturbations of sex hierarchy genes and of DIP-ε change the sex-specific morphologies of fru P1 ∩ DIP-α neurons.


Subject(s)
Cell Adhesion Molecules/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Nerve Tissue Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Adhesion Molecules/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Female , Male , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Sequence Analysis, RNA , Sex Characteristics , Sexual Behavior, Animal , Transcription Factors/genetics
7.
G3 (Bethesda) ; 10(3): 967-983, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31907222

ABSTRACT

Examining cross-tissue interactions is important for understanding physiology and homeostasis. In animals, the female gonad produces signaling molecules that act distally. We examine gene expression in Drosophila melanogaster female head tissues in 1) virgins without a germline compared to virgins with a germline, 2) post-mated females with and without a germline compared to virgins, and 3) post-mated females mated to males with and without a germline compared to virgins. In virgins, the absence of a female germline results in expression changes in genes with known roles in nutrient homeostasis. At one- and three-day(s) post-mating, genes that change expression are enriched with those that function in metabolic pathways, in all conditions. We systematically examine female post-mating impacts on sleep, food preference and re-mating, in the strains and time points used for gene expression analyses and compare to published studies. We show that post-mating, gene expression changes vary by strain, prompting us to examine variation in female re-mating. We perform a genome-wide association study that identifies several DNA polymorphisms, including four in/near Wnt signaling pathway genes. Together, these data reveal how gene expression and behavior in females are influenced by cross-tissue interactions, by examining the impact of mating, fertility, and genotype.


Subject(s)
Drosophila melanogaster/genetics , Fertility/genetics , Reproduction/genetics , Sexual Behavior, Animal , Wnt Signaling Pathway/genetics , Animals , Drosophila melanogaster/physiology , Female , Gene Expression , Genes, Insect , Genome-Wide Association Study , Genotype , INDEL Mutation , Male , Polymorphism, Genetic
8.
Development ; 146(14)2019 07 25.
Article in English | MEDLINE | ID: mdl-31285355

ABSTRACT

The ability of a single genome to produce distinct and often dramatically different male and female forms is one of the wonders of animal development. In Drosophila melanogaster, most sexually dimorphic traits are controlled by sex-specific isoforms of the doublesex (dsx) transcription factor, and dsx expression is mostly limited to cells that give rise to sexually dimorphic traits. However, it is unknown how this mosaic of sexually dimorphic and monomorphic organs arises. Here, we characterize the cis-regulatory sequences that control dsx expression in the foreleg, which contains multiple types of sex-specific sensory organs. We find that separate modular enhancers are responsible for dsx expression in each sexually dimorphic organ. Expression of dsx in the sex comb is co-regulated by two enhancers with distinct spatial and temporal specificities that are separated by a genitalia-specific enhancer. The sex comb-specific enhancer from D. willistoni, a species that primitively lacks sex combs, is not active in the foreleg. Thus, the mosaic of sexually dimorphic and monomorphic organs depends on modular regulation of dsx transcription by dedicated cell type-specific enhancers.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Enhancer Elements, Genetic/physiology , Genitalia/embryology , Genitalia/metabolism , Sex Differentiation/genetics , Animals , Animals, Genetically Modified , DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental , Male , Organ Specificity/genetics , Protein Isoforms/genetics , Protein Isoforms/physiology , Sex Characteristics
9.
Genetics ; 212(2): 365-376, 2019 06.
Article in English | MEDLINE | ID: mdl-31167898

ABSTRACT

Bruce Baker, a preeminent Drosophila geneticist who made fundamental contributions to our understanding of the molecular genetic basis of sex differences, passed away July 1, 2018 at the age of 72. Members of Bruce's laboratory remember him as an intensely dedicated, rigorous, creative, deep-thinking, and fearless scientist. His trainees also remember his strong commitment to teaching students at every level. Bruce's career studying sex differences had three major epochs, where the laboratory was focused on: (1) sex determination and dosage compensation, (2) the development of sex-specific structures, and (3) the molecular genetic basis for sex differences in behavior. Several members of the Baker laboratory have come together to honor Bruce by highlighting some of the laboratory's major scientific contributions in these areas.


Subject(s)
Drosophila/genetics , Genetics/history , Sex Determination Processes/genetics , Animals , Dosage Compensation, Genetic , Evolution, Molecular , History, 20th Century , History, 21st Century , Mentors , Sexual Behavior
10.
G3 (Bethesda) ; 9(8): 2623-2628, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31164388

ABSTRACT

Rodent maternal behaviors are due to the coordinated effects of fluctuating hormones, with their onset triggered by interactions with newborn pups. Previous studies have shown that many genes have changes in expression during peripartum stages. However, it is unclear if there are long-lasting changes in gene expression, well after the performance of maternal behaviors, that could influence physiology and behavior throughout the remaining lifespan. Here, gene expression differences were examined in mouse between age-matched virgin and primiparous females, at least 4 weeks after weaning. Of the five brain regions examined-hypothalamus, hippocampus, cortex, cerebellum, and the amygdala-only the hypothalamus had thousands of genes with significant expression differences. The cerebellum had 130 genes with expression differences, and the other brain regions had no significant changes detected. The expression changes in the hypothalamus include an enrichment of genes that could mediate long-lasting behavioral and physiological changes, given their known roles in parental behavior, including galanin and prolactin receptor.


Subject(s)
Biomarkers , Brain/metabolism , Gene Expression , Animals , Chromosome Mapping , Computational Biology/methods , Female , Gene Expression Profiling , Hippocampus/metabolism , Male , Meta-Analysis as Topic , Mice , Phylogeny , Transcriptome
11.
BMC Genomics ; 20(1): 427, 2019 May 28.
Article in English | MEDLINE | ID: mdl-31138122

ABSTRACT

BACKGROUND: The two Caenorhabditis elegans somatic gonadal precursors (SGPs) are multipotent progenitors that generate all somatic tissues of the adult reproductive system. The sister cells of the SGPs are two head mesodermal cells (hmcs); one hmc dies by programmed cell death and the other terminally differentiates. Thus, a single cell division gives rise to one multipotent progenitor and one differentiated cell with identical lineage histories. We compared the transcriptomes of SGPs and hmcs in order to learn the determinants of multipotency and differentiation in this lineage. RESULTS: We generated a strain that expressed fluorescent markers specifically in SGPs (ehn-3A::tdTomato) and hmcs (bgal-1::GFP). We dissociated cells from animals after the SGP/hmc cell division, but before the SGPs had further divided, and subjected the dissociated cells to fluorescence-activated cell sorting to collect isolated SGPs and hmcs. We analyzed the transcriptomes of these cells and found that 5912 transcripts were significantly differentially expressed, with at least two-fold change in expression, between the two cell types. The hmc-biased genes were enriched with those that are characteristic of neurons. The SGP-biased genes were enriched with those indicative of cell proliferation and development. We assessed the validity of our differentially expressed genes by examining existing reporters for five of the 10 genes with the most significantly biased expression in SGPs and found that two showed expression in SGPs. For one reporter that did not show expression in SGPs, we generated a GFP knock-in using CRISPR/Cas9. This reporter, in the native genomic context, was expressed in SGPs. CONCLUSIONS: We found that the transcriptional profiles of SGPs and hmcs are strikingly different. The hmc-biased genes are enriched with those that encode synaptic transmission machinery, which strongly suggests that it has neuron-like signaling properties. In contrast, the SGP-biased genes are enriched with genes that encode factors involved in transcription and translation, as would be expected from a cell preparing to undergo proliferative divisions. Mediators of multipotency are likely to be among the genes differentially expressed in SGPs.


Subject(s)
Caenorhabditis elegans/genetics , Multipotent Stem Cells/metabolism , Animals , Caenorhabditis elegans/metabolism , Cell Differentiation , Gene Expression Profiling , Gonads/cytology , Mesoderm/cytology , RNA, Messenger/metabolism
12.
BMC Genomics ; 19(1): 893, 2018 Dec 10.
Article in English | MEDLINE | ID: mdl-30526477

ABSTRACT

BACKGROUND: The core functions of the insulin/insulin-like signaling and target of rapamycin (IIS/TOR) pathway are nutrient sensing, energy homeostasis, growth, and regulation of stress responses. This pathway is also known to interact directly and indirectly with the sex determination regulatory hierarchy. The IIS/TOR pathway plays a role in directing sexually dimorphic traits, including dimorphism of growth, metabolism, stress and behavior. Previous studies of sexually dimorphic gene expression in the adult head, which includes both nervous system and endocrine tissues, have revealed variation in sex-differential expression, depending in part on genotype and environment. To understand the degree to which the environmentally responsive insulin signaling pathway contributes to sexual dimorphism of gene expression, we examined the effect of perturbation of the pathway on gene expression in male and female Drosophila heads. RESULTS: Our data reveal a large effect of insulin signaling on gene expression, with greater than 50% of genes examined changing expression. Males and females have a shared gene expression response to knock-down of InR function, with significant enrichment for pathways involved in metabolism. Perturbation of insulin signaling has a greater impact on gene expression in males, with more genes changing expression and with gene expression differences of larger magnitude. Primarily as a consequence of the response in males, we find that reduced insulin signaling results in a striking increase in sex-differential expression. This includes sex-differences in expression of immune, defense and stress response genes, genes involved in modulating reproductive behavior, genes linking insulin signaling and ageing, and in the insulin signaling pathway itself. CONCLUSIONS: Our results demonstrate that perturbation of insulin signaling results in thousands of genes displaying sex differences in expression that are not differentially expressed in control conditions. Thus, insulin signaling may play a role in variability of somatic, sex-differential expression. The finding that perturbation of the IIS/TOR pathway results in an altered landscape of sex-differential expression suggests a role of insulin signaling in the physiological underpinnings of trade-offs, sexual conflict and sex differences in expression variability.


Subject(s)
Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Insulin/metabolism , Sex Characteristics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Exons/genetics , Female , Male , Receptor, Insulin/metabolism
13.
G3 (Bethesda) ; 6(8): 2455-65, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27247289

ABSTRACT

Male and female reproductive behaviors in Drosophila melanogaster are vastly different, but neurons that express sex-specifically spliced fruitless transcripts (fru P1) underlie these behaviors in both sexes. How this set of neurons can generate such different behaviors between the two sexes is an unresolved question. A particular challenge is that fru P1-expressing neurons comprise only 2-5% of the adult nervous system, and so studies of adult head tissue or whole brain may not reveal crucial differences. Translating Ribosome Affinity Purification (TRAP) identifies the actively translated pool of mRNAs from fru P1-expressing neurons, allowing a sensitive, cell-type-specific assay. We find four times more male-biased than female-biased genes in TRAP mRNAs from fru P1-expressing neurons. This suggests a potential mechanism to generate dimorphism in behavior. The male-biased genes may direct male behaviors by establishing cell fate in a similar context of gene expression observed in females. These results suggest a possible global mechanism for how distinct behaviors can arise from a shared set of neurons.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Nerve Tissue Proteins/genetics , Reproduction/genetics , Sexual Behavior, Animal , Transcription Factors/genetics , Animals , Drosophila melanogaster/growth & development , Female , Gene Expression Regulation, Developmental , Male , Neurons/metabolism , Sex Characteristics
14.
Dev Cell ; 37(6): 486-8, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27326925

ABSTRACT

During courtship, Drosophila melanogaster males sing to females a song composed of rhythmic pulses and sine song. In this issue of Developmental Cell, Shirangi et al. (2016) show that a cluster of doublesex-expressing neurons directs the production of the sine song component through functional linkages to wing motoneurons.


Subject(s)
Courtship , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Vocalization, Animal/physiology , Animals , Female , Male , Models, Biological , Neurons/metabolism
15.
Evol Dev ; 18(3): 201-9, 2016 05.
Article in English | MEDLINE | ID: mdl-27161950

ABSTRACT

Animal development is the product of distinct components and interactions-genes, regulatory networks, and cells-and it exhibits emergent properties that cannot be inferred from the components in isolation. Often the focus is on the genotype-to-phenotype map, overlooking the process of development that turns one into the other. We propose a move toward micro-evolutionary analysis of development, incorporating new tools that enable cell type resolution and single-cell microscopy. Using the sex determination pathway in Drosophila to illustrate potential avenues of research, we highlight some of the questions that these emerging technologies can address. For example, they provide an unprecedented opportunity to study heterogeneity within cell populations, and the potential to add the dimension of time to gene regulatory network analysis. Challenges still remain in developing methods to analyze this data and to increase the throughput. However this line of research has the potential to bridge the gaps between previously more disparate fields, such as population genetics and development, opening up new avenues of research.


Subject(s)
Biological Evolution , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Gene Regulatory Networks , Animals , Brain , Gene Expression , Sex Determination Processes
16.
G3 (Bethesda) ; 6(7): 1799-808, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27172187

ABSTRACT

Sex differences in gene expression have been widely studied in Drosophila melanogaster Sex differences vary across strains, but many molecular studies focus on only a single strain, or on genes that show sexually dimorphic expression in many strains. How extensive variability is and whether this variability occurs among genes regulated by sex determination hierarchy terminal transcription factors is unknown. To address these questions, we examine differences in sexually dimorphic gene expression between two strains in Drosophila adult head tissues. We also examine gene expression in doublesex (dsx) mutant strains to determine which sex-differentially expressed genes are regulated by DSX, and the mode by which DSX regulates expression. We find substantial variation in sex-differential expression. The sets of genes with sexually dimorphic expression in each strain show little overlap. The prevalence of different DSX regulatory modes also varies between the two strains. Neither the patterns of DSX DNA occupancy, nor mode of DSX regulation explain why some genes show consistent sex-differential expression across strains. We find that the genes identified as regulated by DSX in this study are enriched with known sites of DSX DNA occupancy. Finally, we find that sex-differentially expressed genes and genes regulated by DSX are highly enriched on the fourth chromosome. These results provide insights into a more complete pool of potential DSX targets, as well as revealing the molecular flexibility of DSX regulation.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Sex Characteristics , Sex Determination Processes , Transcriptome , Animals , Chromosome Mapping , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Female , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Interaction Mapping , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
17.
J Comp Neurol ; 524(13): 2696-710, 2016 09 01.
Article in English | MEDLINE | ID: mdl-26917114

ABSTRACT

Identifying sex differences in gene expression within the brain is critical for determining why multiple neurological and behavioral disorders differentially affect males and females. Several disorders are more common or severe in males (e.g., autism and schizophrenia) or in females (e.g., Alzheimer's disease and depression). We analyzed transcriptomic data from the mouse hippocampus of six inbred strains (129S1/SvImJ, A/J, C57BL/6J, DBA/1J, DBA/2J, and PWD/Ph) to provide a perspective on differences between male and female gene expression. Our data show that 1) gene expression differences in males vs. females varies substantially across the strains, 2) only a few genes are differentially expressed across all of the strains (termed core genes), and 3) >2,600 genes differ in the individual strain comparisons (termed noncore genes). We found that DBA/2J uniquely has a substantial majority (89%) of differentially expressed genes (DEGs) that are more highly expressed in females than in males (female-biased); 129/SvImJ has a majority (69%) of DEGs that are more highly expressed in males. To gain insight into the function of the DEGs, we examined gene ontology and pathway and phenotype enrichment and found significant enrichment in phenotypes related to abnormal nervous system morphology and physiology, among others. In addition, several pathways are enriched significantly, including Alzheimer's disease (AD), with 32 genes implicated in AD, eight of which are male-biased. Three of the male-biased genes have been implicated in a neuroprotective role in AD. Our transcriptomic data provide new insight into the possible genetic bases for sex-specific susceptibility and severity of brain disorders. J. Comp. Neurol. 524:2696-2710, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Gene Expression Profiling/methods , Hippocampus/physiology , Nervous System Diseases/genetics , Severity of Illness Index , Sex Characteristics , Animals , Female , Gene Regulatory Networks/genetics , Hippocampus/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Nervous System Diseases/pathology , Species Specificity
18.
G3 (Bethesda) ; 6(1): 221-33, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26596646

ABSTRACT

The developmental transition to motherhood requires gene expression changes that alter the brain to drive the female to perform maternal behaviors. We broadly examined the global transcriptional response in the mouse maternal brain, by examining four brain regions: hypothalamus, hippocampus, neocortex, and cerebellum, in virgin females, two pregnancy time points, and three postpartum time points. We find that overall there are hundreds of differentially expressed genes, but each brain region and time point shows a unique molecular signature, with only 49 genes differentially expressed in all four regions. Interestingly, a set of "early-response genes" is repressed in all brain regions during pregnancy and postpartum stages. Several genes previously implicated in underlying postpartum depression change expression. This study serves as an atlas of gene expression changes in the maternal brain, with the results demonstrating that pregnancy, parturition, and postpartum maternal experience substantially impact diverse brain regions.


Subject(s)
Brain/metabolism , Gene Expression Regulation , Postpartum Period , Animals , Behavior, Animal , Cluster Analysis , Computational Biology/methods , Depression, Postpartum/genetics , Female , Gene Expression Profiling , Gene Ontology , High-Throughput Nucleotide Sequencing , Humans , Male , Mice , Phenotype , Pregnancy , Transcriptome
19.
BMC Syst Biol ; 9: 53, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26335107

ABSTRACT

BACKGROUND: The Drosophila sex determination hierarchy is a classic example of a transcriptional regulatory hierarchy, with sex-specific isoforms regulating morphology and behavior. We use a structural equation modeling approach, leveraging natural genetic variation from two studies on Drosophila female head tissues--DSPR collection (596 F1-hybrids from crosses between DSPR sub-populations) and CEGS population (75 F1-hybrids from crosses between DGRP/Winters lines to a reference strain w1118)--to expand understanding of the sex hierarchy gene regulatory network (GRN). This approach is completely generalizable to any natural population, including humans. RESULTS: We expanded the sex hierarchy GRN adding novel links among genes, including a link from fruitless (fru) to Sex-lethal (Sxl) identified in both populations. This link is further supported by the presence of fru binding sites in the Sxl locus. 754 candidate genes were added to the pathway, including the splicing factors male-specific lethal 2 and Rm62 as downstream targets of Sxl which are well-supported links in males. Independent studies of doublesex and transformer mutants support many additions, including evidence for a link between the sex hierarchy and metabolism, via Insulin-like receptor. CONCLUSIONS: The genes added in the CEGS population were enriched for genes with sex-biased splicing and components of the spliceosome. A common goal of molecular biologists is to expand understanding about regulatory interactions among genes. Using natural alleles we can not only identify novel relationships, but using supervised approaches can order genes into a regulatory hierarchy. Combining these results with independent large effect mutation studies, allows clear candidates for detailed molecular follow-up to emerge.


Subject(s)
Computational Biology/methods , Drosophila melanogaster/genetics , Gene Regulatory Networks , Models, Genetic , Sex Determination Processes/genetics , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Female , Male , Nuclear Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Supervised Machine Learning , Unsupervised Machine Learning
SELECTION OF CITATIONS
SEARCH DETAIL
...