Abstract
The transcriptome of mammalian tissues differs between males and females, and these differences can change across the lifespan, likely regulating known sexual dimorphisms in disease prevalence and severity. Cataract, the most prevalent disease of the ocular lens, occurs at similar rates in young individuals, but its incidence is elevated in older women compared to men of the same age. However, the influence of sex on the lens transcriptome was unknown. RNAseq based transcriptomic profiling of young adult C57BL/6J mouse lens epithelial and fiber cells revealed that few genes are differentially expressed between the sexes. In contrast, lens cells from aged (24 month old) male and female C57BL/6J mice differentially expressed many genes, including several whose expression is lens preferred. . Like cataracts, posterior capsular opacification (PCO), a major sequela of cataract surgery, may also be more prevalent in women. Lens epithelial cells isolated from mouse eyes 24 hours after lens fiber cell removal exhibited numerous transcriptomic differences between the sexes, including genes implicated in complement cascades and extracellular matrix regulation, and these differences are much more pronounced in aged mice than in young mice. These results provide an unbiased basis for future studies on how sex affects the lens response to aging, cataract development, and cataract surgery.
Introduction
Biological sex in vertebrates is commonly recognized as the set of male or female traits necessary for an organism to successfully contribute to sexual reproduction. In mammals, this is conferred primarily by the organism’s sex chromosome complement, with female primary sexual characteristics considered as the “default” state. Males form when genes present on the Y chromosome drive testes differentiation from the genital ridge. The resulting Leydig cell differentiation leads to the fetal testosterone production necessary for the development of the male genitalia (Greenfield, 2015; Zhao and Yao, 2019). This concept was further bolstered by the realization that females, which carry two copies of the X chromosome, randomly inactivate one copy via the action of the long non-coding RNA Xist, yielding males and females who have similar X chromosome gene dosage (Arnold, 2017).
However, sex affects numerous aspects of biology besides those directly relevant to reproduction, as exemplified by the wide variety of human diseases for which disease incidence, severity, or presentation differs greatly between sexes (Mauvais-Jarvis et al., 2020). These differences, along with sexually dimorphic aging mechanisms, likely contribute to the longer life expectancy of women compared to men (Sampathkumar et al., 2020). However, in contrast to this, epidemiological studies have consistently found that post-menopausal women have a higher risk of cataract than age-matched men, although the cataract rates in younger people are similar in both sexes (Zetterberg and Celojevic, 2015). Further, several epidemiological studies have suggested that women have a significantly higher risk of developing posterior capsular opacification (PCO) after cataract surgery than men (Congdon et al., 2008; Fong et al., 2014; Lee et al., 2016), although other studies do not support this correlation (Tokko et al., 2019).
Most experimental investigations into the increased risk of cataract in post-menopausal women have focused on a possible protective effect of estrogen on cataract development (Zetterberg and Celojevic, 2015). The prevalent hypothesis in the literature suggests that the withdrawal of estrogen at menopause results in cataract due to a “rebound” effect on the lens (Zetterberg and Celojevic, 2015). This postulate is supported by epidemiological studies finding a correlation between hormonal replacement therapy of menopausal women and protection from cataract (Noran et al., 2007; Younan et al., 2002), while treatment of breast cancer patients with the anti-estrogen, tamoxifen, increases cataract risk (Gorin et al., 1998; Nelson et al., 2019).
In other tissues, sex confers more differences on cells than can be explained by just differential exposure to sex hormones, including changes in DNA methylation (Hodes et al., 2017) and the expression of numerous autosomal genes (Arnold, 2019), even prior to embryonic sex determination (Lowe et al., 2015). However, no studies have investigated how sex affects global gene expression in the lens, and it is unknown whether any sex differences vary during aging. We used RNAseq to perform transcriptome profiling of lens cells obtained from inbred C57BL/6J strain mice to discover the effect of age on the lens epithelial and fiber cell transcriptomes, and the transcriptional response of lens epithelial cells to a surgery that models lens epithelium injury during cataract surgery (Faranda et al, 2021). As that study was performed on samples from both sexes, here, we reanalyze that dataset to elucidate how the global transcriptome of lens epithelial and fiber cells isolated from young (3 months) and old (24 month) old C57BL/6J strain mice differ by sex and to investigate how sex and age affect the response of lens epithelial cells to lens fiber cell removal performed to model modern cataract surgery. As the National Institutes of Health of the United States has mandated that research studies should include animals from both sexes, and consider the possibility that sex is a relevant biological variable in biomedical research (Woitowich et al., 2020), this study will provide important baseline information for the field of lens and cataract research.
Materials and methods:
Mice
All studies using mice comply with the Association for Research in Vision and Ophthalmology Statement on the Use of Animals in Vision Research and were approved by the University of Delaware Institutional Animal Care and Use Committee. Twenty four month old C57BL/6NIA mice (10 males and 10 females) were obtained from the National Institute on Aging Biological Resources Colony in October of 2018. These animals are derived from C57BL/6J foundation stock obtained from the Jackson Laboratory in 2016. Ten week old C57BL/6J mice (10 males and 10 females, Stock # 000664) were obtained from the Jackson Laboratory in October of 2018. In both cases, animals were housed at the University of Delaware animal facility under a 14/10 hour light-dark cycle for two weeks prior to tissue isolation. The eyes from all mice used in this study were of normal size and did not manifest signs of the sporadic eye defects that have been reported in this strain (Smith et al., 1994).
Mouse cataract surgery model and tissue isolation
Surgical removal of lens fiber cells was performed on adult mice to mimic human cataract surgery as previously described (Desai et al., 2010; Manthey et al., 2014b) following procedures first reported by Call et al. (Call et al., 2004). Briefly, two weeks after arrival at the University of Delaware, mice were anesthetized following iris dilation, a central corneal incision made, and the entire lens fiber cell mass was removed from one eye by a sharp forceps, leaving behind an intact lens capsule. The cornea was sutured and the eye restored to normal shape with balanced saline solution. Twenty four hours later, mice were re- anesthetized, and the surgery repeated on the other eye. Mice were then immediately sacrificed and lens capsular bags isolated by dissection.
For RNA sequencing, lens capsular bags from either 24 hours post cataract surgery (PCS) or zero hours PCS were pooled from five animals to make a single biological replicate, while lens fiber masses from two independent animals were pooled per replicate. Two biological replicates were created for each condition (male and female samples from 3 month old versus 24 month old animals at zero hours PCS, 24 hours PCS, and lens fiber cells), and flash frozen on dry ice. RNA was isolated from these samples, RNA libraries prepared using the SMARTer® Stranded Total RNA-Seq Kit-Pico Input Mammalian (Takara Bio USA, Inc., Mountain View, CA, USA) and sequenced by DNA Link, USA (901 Morena Blvd. Ste 730 San Diego CA 92117, USA) on a NovaSeq 6000 (San Diego, CA, USA). Read pairs (101 basepairs, bidirectional) were aligned to the Ensembl primary assembly of the mouse GRCm38 genome (Yates et al., 2020) using Hisat2 with its default parameters (Kim et al., 2019). Read pairs aligning to genomic features in the Ensembl Mouse version 101 GTF file were quantified as gene level counts, using HTSeq-Count in union mode (Anders et al., 2014). Length normalized abundance estimates (Fragments per Kilobase-Million (FPKM)) were calculated from gene level counts using the total length of all known exons for a given gene, after merging overlapping exons.
Samples were partitioned for TMM (Trimmed Median of Means) scaling (Robinson and Oshlack, 2010) and differential expression analyses were performed based on the objective of a particular contrast. For contrasts evaluating differences between epithelial cells and fiber cells, and sex or age effects in un-injured tissues, all samples collected at 0 hours post-surgery were grouped together. For contrasts evaluating LEC injury responses, all LECs samples were grouped together.
The “exactTest” method from the edgeR statistical package (version 3.30.3) was used to estimate the magnitude and statistical significance of differential gene expression, with robust dispersion estimates (Phipson et al., 2016; Robinson et al., 2010). Genes with at least 10 mapped reads in at least two samples were considered to have “detectable” levels of expression. Genes failing “detectable” criteria were removed prior to running the “exactTest”, using edgeR’s “filterByExpr” function (Chen et al., 2016).
Biologically significant differentially expressed genes (DEGs) were defined as those exhibiting a statistically significant difference in expression using Storey’s Q value to adjust for False Discovery Rate (FDR ≤ 0.05; (Storey and Tibshirani, 2003)), a difference in expression level greater than 2 FPKM between conditions, Fold Change (FC) greater than 2 in either the positive or negative direction, and expressed at a level greater than 2 FPKM. (Manthey et al., 2014a).
Pathway analyses
Pathway analysis was performed on all statistically significant DEGs defined as those exhibiting a fold change ≥ |2| and FDR < 0.05 using iPathwayGuide (Advaita Bioinformatics, Plymouth Michigan, USA). This software package uses Impact Analysis, an approach that considers the directed interactions of a DEG within a given pathway (as defined by the Kyoto Encyclopedia of Genes and Genomes, KEGG, (Kanehisa et al., 2017), Release 96.0+/11-21, Nov 20), and also whether more pathway participants are observed in the DEG list than would be expected by chance (Ahsan and Draghici, 2017; Draghici et al., 2007; Tarca et al., 2009). Gene ontology comparisons were made against the October 14, 2020 release of the Gene Ontology Consortium database
Lens Preferred Genes in the iSyte database:
The Integrated systems tool for eye gene discovery (iSyTe) database (Kakrana et al., 2018) uses the principle of “whole body subtraction” to identify genes with a significant difference in expression between the lens, and the rest of the body. Genes expressed at higher levels in the lens vs non-lens tissues are described as “lens-preferred”, and are expected to contribute to the lens phenotype. In the current study, DEGs identified in sex and age dependent contrasts were compared to the 412 genes that iSyTe considers “highly lens preferred” at postnatal day 56.
Results
We studied how aging affects the lens transcriptome by performing RNAseq analyses on lens cells isolated from 3 and 24 month old inbred C57BL/6J strain mice housed under standard laboratory conditions (Faranda et al., 2021) as this minimized potential variability driven by differences in genetic background and environment (Ackert-Bicknell et al., 2015). During the design of this study, we included equal numbers of animals from both sexes as mandated by NIH guidelines (Woitowich et al., 2020), which led us to reexamine these data (Gene Expression Omnibus Accession number GSE166619) to discover how sex affects the lens transcriptome. Principal component analysis (PCA) and a Spearman’s Rho correlation matrix revealed that cell type (epithelium versus fibers) was the largest influence on transcriptomic differences in this study, followed by the effect of injury on the lens epithelial cell (LEC) transcriptome, followed by age. The greatest variation between replicates was seen in samples from injured lens samples suggesting some sample to sample variability in the injury response at 24 hours post surgery. (Supplemental Figure 1). However, as expected for inbred mice housed under controlled conditions, Spearman’s Rho correlation plots comparing the global levels of gene expression between sex specific biological replicates strongly correlate (Supplemental figure 2) although, sex appears to have much less influence on the global lens transcriptome than cell type, injury status, or age (Supplemental figure 1).
The effect of sex on the transcriptome of the young adult lens epithelium
Consistent with the PCA analysis, comparison between the transcriptome of young adult male and female mouse lens epithelial cells revealed only five differentially expressed genes (DEGs) that met our previously developed criteria for “biologically significant” changes that could plausibly affect lens cells (at least 2 fold change in expression level, FDR corrected p value ≤ 0.05, gene expressed at least 2 FPKM in at least one condition (Manthey et al., 2014a)). Of these, three were genes residing on sex chromosomes, while the other two are predicted genes of unknown function that reside on autosomes. All three genes residing on sex chromosomes exhibit sex-dependent expression in other mouse tissues as well based on comparisons with the Sex associated genes data base (SAGD (Shi et al., 2019)), while neither autosomal gene has been reported to be sex-dependent in SAGD (Table 1). Of these genes, only Xist exhibited “lens preferred” expression in 56 day postnatal LECs based on comparisons with the Integrated systems tool for eye gene discovery (iSyTe) database (Kakrana et al., 2018) which likely reflects the sex of the tissue used to create this resource. Too few genes exhibited significant differential expression between male and female young LECs to perform Adviata ipathway guide analyses (Ahsan and Draghici, 2017) on this gene list.
Table 1.
DEGs between young male and female lens epithelial cells. Bold denotes genes that have been previously reported to exhibit sex dependent expression in adult mouse tissues based on comparisons with SAGD (Shi et al., 2019). Italics denotes genes that exhibit lens specific expression in postnatal day 56 mouse lenses according to the iSyTe database (Kakrana et al., 2018).
SYMBOL | DESCRIPTION | Chromosome | Fold Change | FDR | Male FPKM | Female FPKM |
---|---|---|---|---|---|---|
Xist | inactive X specific transcripts | X | 2756.3 | 1.7E-91 | 0.01 | 28.3 |
Ddx3y | DEAD (Asp-Glu-Ala-Asp) box polypeptide 3, Y-linked | Y | −8455.0 | 2.8E-89 | 6.3 | 0.0 |
Eif2s3y | eukaryotic translation initiation factor 2, subunit 3, Y-linked | Y | −4692.1 | 2.2E-25 | 4.4 | 0.0 |
Gm10800 | predicted gene 10800 | 2 | 2114.4 | 3.1E-8 | 0.00 | 12.4 |
Gm42972 | predicted gene 42972 | 3 | −2.8 | 3.5E-2 | 4.9 | 1.8 |
The effect of sex on the response of young adult lens epithelial cells to lens fiber cell removal
Comparison between the transcriptome of young adult male and female mouse lens epithelial cells isolated 24 hours after lens fiber cell removal to model cataract surgery (Jiang et al., 2018; Manthey et al., 2014b) revealed only ten biologically significant DEGs; six are located on sex chromosomes, four on autosomes. Of these, only Xist exhibited lens-preferred expression in adults based on comparisons with the iSyTe database and only five genes (all located on sex chromosomes) exhibit sexually dimorphic gene expression in adult mice in other tissues based on SAGD (Table 2). We were unable to perform iPathway guide analyses of these DEGs as too few genes were identified for the underlying algorithm to detect enriched pathways.
Table 2.
DEGs between young male and female lens epithelial cells at 24 hours PCS. Bold denotes genes that have been previously reported to exhibit sex dependent expression in adult mouse tissues based on comparisons with SAGD (Shi et al., 2019). Italics denotes genes that exhibit lens preferred expression in postnatal day 56 mouse lenses according to the iSyTe database (Kakrana et al., 2018).
SYMBOL | DESCRIPTION | Chromosome | Fold Change | FDR | Male FPKM | Female FPKM |
---|---|---|---|---|---|---|
Xist | inactive X specific transcripts | X | 1672.4 | 2.63E-3 | 0.0 | 49.5 |
Gm13375 | predicted gene 13375 | 2 | 16.2 | 4.91E-2 | 0.2 | 2.5 |
Eif2s3x | eukaryotic translation initiation factor 2, subunit 3, X-linked [ | X | 2.7 | 4.47E-3 | 9.9 | 27.2 |
Serping1 | serine (or cysteine) peptidase inhibitor, clade G, member 1 | 2 | 2.4 | 4.38E-2 | 7.8 | 18.7 |
G0s2 | G0/G1 switch gene 2 | 1 | −3.1 | 4.38E-2 | 35.6 | 11.6 |
Ccdc88b | coiled-coil domain containing 88B | 19 | −3.6 | 1.01E-2 | 8.1 | 2.2 |
Uty | ubiquitously transcribed tetratricopeptide repeat gene, Y chromosome | Y | −3158.4 | 2.27E-34 | 2.1 | 0.0 |
Eif2s3y | eukaryotic translation initiation factor 2, subunit 3, Y-linked | Y | −3449.3 | 2.17E-38 | 5.4 | 0.0 |
Kdm5d | lysine (K)-specific demethylase 5D | Y | −5243.9 | 7.43E-44 | 2.2 | 0.0 |
Ddx3y | DEAD (Asp-Glu-Ala-Asp) box polypeptide 3, Y-linked | Y | −6043.4 | 2.11E-59 | 7.5 | 0.0 |
The effect of sex on the young adult lens fiber cell transcriptome
Comparison between the transcriptome of young adult male and female mouse lens fiber cells revealed that 49 genes met the criteria for “biologically significant” differential expression between the sexes, with 28 genes expressed at higher levels in females and 21 expressed at higher levels in males. Of these, five DEGs reside on sex chromosomes while the remainder are autosomal (Table 3). Advaita iPathway guide (Ahsan and Draghici, 2017) analysis of the 191 DEGs meeting statistical criteria for differential expression (FDR ≤ 0.05; absolute fold change > 2; 191 genes total) did not detect any ontology terms or KEGG pathways suggesting common regulated mechanisms.
Table 3.
DEGs between young male and female lens fiber cells. Bold denotes genes that have been previously reported to exhibit sex dependent expression in adult mouse tissues based on comparisons with SAGD (Shi et al., 2019). Italics denotes genes that exhibit lens preferred expression in postnatal day 56 mouse lenses according to the iSyTe database (Kakrana et al., 2018).
SYMBOL | DESCRIPTION | Chromosome | Fold Change | FDR | Male FPKM | Female FPKM |
---|---|---|---|---|---|---|
Xist | inactive X specific transcripts | X | 312.7 | 9.72E-48 | 0.0 | 10.1 |
Adrb2 | adrenergic receptor, beta 2 | 18 | 173.0 | 7.14E-3 | 0.0 | 2.3 |
Mrpl22 | mitochondrial ribosomal protein L22 | 11 | 16.5 | 9.66E-3 | 0.2 | 4.1 |
Tm7sf3 | transmembrane 7 superfamily member 3 | 6 | 13.1 | 3.63E-8 | 0.3 | 4.3 |
Selenon | selenoprotein N | 4 | 6.3 | 8.76E-7 | 0.7 | 4.3 |
Gm10557 | predicted gene 10557 | 18 | 5.9 | 6.90E-3 | 1.9 | 11.8 |
Sdhc | succinate dehydrogenase complex, subunit C | 1 | 5.8 | 5.00E-4 | 0.6 | 3.6 |
Mfsd1 | major facilitator superfamily domain containing 1 | 3 | 5.7 | 5.01E-3 | 0.5 | 2.9 |
Olfml3 | olfactomedin-like 3 | 3 | 5.0 | 1.31E-5 | 0.7 | 3.3 |
Ppp3cc | protein phosphatase 3, catalytic subunit, gamma isoform | 14 | 4.7 | 4.52E-2 | 0.6 | 2.7 |
Rab39b | RAB39B, member RAS oncogene family | X | 4.1 | 2.43E-2 | 0.8 | 3.4 |
Rassf7 | Ras association (RalGDS/AF-6) domain family (N-terminal) 7 | 7 | 3.8 | 3.79E-2 | 1.2 | 4.8 |
Samd1 | sterile alpha motif domain containing 1 | 8 | 3.7 | 1.70E-3 | 1.6 | 5.9 |
Ahcy | S-adenosylhomocysteine hydrolase | 2 | 3.5 | 1.14E-2 | 2.4 | 8.4 |
Armc10 | armadillo repeat containing 10 | 5 | 3.3 | 2.62E-2 | 1.2 | 3.9 |
Bnip2 | BCL2/adenovirus E1B interacting protein 2 | 9 | 2.8 | 2.48E-2 | 1.3 | 3.6 |
Aldh3a1 | aldehyde dehydrogenase family 3, subfamily A1 | 11 | 2.8 | 7.19E-4 | 5.5 | 15.6 |
Mapkapk2 | MAP kinase-activated protein kinase 2 | 1 | 2.7 | 2.01E-2 | 1.2 | 3.4 |
Rbm7 | RNA binding motif protein 7 | 9 | 2.6 | 2.35E-2 | 1.4 | 3.6 |
Ptpn2 | protein tyrosine phosphatase, non-receptor type 2 | 18 | 2.6 | 2.69E-2 | 2.6 | 6.5 |
Epg5 | ectopic P-granules autophagy protein 5 homolog (C. elegans) | 18 | 2.5 | 1.29E-3 | 1.4 | 3.5 |
Retreg1 | reticulophagy regulator 1 | 15 | 2.5 | 5.94E-3 | 2.4 | 6.0 |
Flcn | folliculin | 11 | 2.3 | 2.62E-2 | 1.8 | 4.2 |
Acat1 | acetyl-Coenzyme A acetyltransferase 1 | 9 | 2.3 | 4.15E-2 | 2.3 | 5.4 |
Gstm1 | glutathione S-transferase, mu 1 | 3 | 2.3 | 6.70E-4 | 15.1 | 34.7 |
2410004B18Rik | RIKEN cDNA 2410004B18 gene | 3 | 2.2 | 6.39E-3 | 8.1 | 18.2 |
Zc3h4 | zinc finger CCCH-type containing 4 | 7 | 2.1 | 5.86E-4 | 2.3 | 4.9 |
Exoc3 | exocyst complex component 3 | 13 | 2.0 | 1.14E-2 | 3.7 | 7.5 |
Arpc5 | actin related protein 2/3 complex, subunit 5 | 1 | −2.0 | 3.02E-2 | 15.3 | 7.6 |
Hmgb3 | high mobility group box 3 | X | −2.1 | 5.94E-3 | 25.3 | 12.3 |
Cxxc1 | CXXC finger 1 (PHD domain) | 18 | −2.1 | 1.81E-2 | 8.4 | 4.0 |
Mtmr4 | myotubularin related protein 4 | 11 | −2.2 | 3.79E-2 | 5.3 | 2.4 |
Arhgef19 | Rho guanine nucleotide exchange factor (GEF) 19 | 4 | −2.2 | 2.46E-2 | 3.7 | 1.7 |
Ahdc1 | AT hook, DNA binding motif, containing 1 | 4 | −2.3 | 5.53E-4 | 4.1 | 1.8 |
Oplah | 5-oxoprolinase (ATP-hydrolysing) | 15 | −2.3 | 1.14E-2 | 3.8 | 1.7 |
Tm9sf1 | transmembrane 9 superfamily member 1 | 14 | −2.7 | 1.79E-2 | 3.5 | 1.3 |
Zdhhc4 | zinc finger, DHHC domain containing 4 | 5 | −2.7 | 3.85E-2 | 7.1 | 2.6 |
Susd4 | sushi domain containing 4 | 1 | −2.9 | 2.83E-2 | 5.0 | 1.7 |
Spsb3 | splA/ryanodine receptor domain and SOCS box containing 3 | 17 | −2.9 | 4.52E-2 | 6.3 | 2.1 |
Pygm | muscle glycogen phosphorylase | 19 | −3.1 | 7.34E-3 | 4.6 | 1.5 |
Gm14322 | predicted gene 14322 | 2 | −3.2 | 3.71E-2 | 13.3 | 4.1 |
Map2k1 | mitogen-activated protein kinase kinase 1 | 9 | −3.3 | 1.15E-4 | 6.2 | 1.9 |
Cntfr | ciliary neurotrophic factor receptor | 4 | −3.8 | 4.15E-2 | 3.2 | 0.8 |
Yipf2 | Yip1 domain family, member 2 | 9 | −6.4 | 5.09E-3 | 4.6 | 0.7 |
S100a1 | S100 calcium binding protein A1 | 3 | −17.3 | 1.02E-2 | 6.7 | 0.3 |
Tmub1 | transmembrane and ubiquitin-like domain containing 1 | 5 | −18.5 | 1.04E-3 | 2.6 | 0.1 |
A930036I15Rik | RIKEN cDNA A930036I15 gene | 3 | −81.7 | 3.91E-2 | 2.2 | 0.0 |
Ddx3y | DEAD (Asp-Glu-Ala-Asp) box polypeptide 3, Y-linked | Y | −335.4 | 1.68E-43 | 9.6 | 0.0 |
Eif2s3y | eukaryotic translation initiation factor 2, subunit 3, Y-linked | Y | −570.3 | 7.04E-11 | 3.9 | 0.0 |
Analysis of these DEGs using iSyte (Kakrana et al., 2018) revealed that only three genes differentially expressed between male and female young lens fibers (Gstm1, Aldh3a1, and Pygm) exhibited lens preferred expression in 56-day old whole lenses. Four DEGs located on sex chromosomes and five autosomal DEGs were previously found to exhibit sexually dimorphic expression in other mouse tissues based on comparisons with SAGD (Table 3).
The effect of sex on the aged lens epithelial cell transcriptome
Comparison between the transcriptome of aged male and female mouse LECs revealed 37 DEGs between the sexes, with 24 genes expressed at higher levels in females and 13 expressed at higher levels in males. Of these, three DEGs reside on sex chromosomes while the remainder are autosomal (Table 4). Three DEGs located on sex chromosomes and nine autosomal DEGs were previously found to exhibit sex dependent expression in other mouse tissues based on comparisons with SAGD (Table 4). Analysis of all DEGs with a statistically significant difference in expression (FDR < 0.05, absolute fold change ≥ 2, 74 genes total) for impacted pathways or enriched gene ontology (GO) terms using iPathway guide (Ahsan and Draghici, 2017) revealed that “lens development in camera type eye” (FDR corrected p value 5 X10−7) and structural constituent of the eye lens (FDR 2.3 X10−11) as enriched GO terms.-As in young LECs, KEGG Pathway analysis was not informative.
Table 4.
DEGs between aged male and female lens epithelial cells. Bold denotes genes that have been previously reported to exhibit sex dependent expression in adult mouse tissues based on comparisons with SAGD (Shi et al., 2019). Italics denotes genes that exhibit lens preferred expression in postnatal day 56 mouse lenses according to the iSyTe database (Kakrana et al., 2018).
SYMBOL | DESCRIPTION | Chromosome | Fold Change | FDR | Male FPKM | Female FPKM |
---|---|---|---|---|---|---|
Xist | inactive X specific transcripts | X | 1932.6 | 3.61E-92 | 0.0 | 34.6 |
Crygd | crystallin, gamma D | 1 | 60.3 | 2.36E-2 | 0.0 | 2.8 |
Gm49958 | predicted gene, 49958 | 17 | 7.9 | 6.62E-5 | 0.3 | 2.5 |
Rgs4 | regulator of G-protein signaling 4 | 1 | 5.2 | 1.29E-2 | 0.6 | 3.3 |
Lgsn | lengsin, lens protein with glutamine synthetase domain | 1 | 4.5 | 4.79E-3 | 19.9 | 88.6 |
Pip5kl1 | phosphatidylinositol-4-phosphate 5-kinase-like 1 | 2 | 3.6 | 1.18E-3 | 1.1 | 3.8 |
Gm42808 | predicted gene 42808 | 5 | 3.5 | 5.55E-3 | 10.6 | 36.9 |
Fabp5 | fatty acid binding protein 5, epidermal | 3 | 3.3 | 1.18E-2 | 14.8 | 48.1 |
Acta2 | actin, alpha 2, smooth muscle, aorta | 19 | 3.3 | 4.76E-9 | 10.8 | 35.1 |
Cryba4 | crystallin, beta A4 | 5 | 3.2 | 5.90E-3 | 179.0 | 581.3 |
Rny1 | RNA, Y1 small cytoplasmic, Ro-associated | 6 | 3.2 | 3.16E-2 | 10.8 | 34.7 |
Pstpip2 | proline-serine-threonine phosphatase-interacting protein 2 | 18 | 2.8 | 1.11E-3 | 1.3 | 3.8 |
Mboat1 | membrane bound O-acyltransferase domain containing 1 | 13 | 2.8 | 2.73E-9 | 3.2 | 8.9 |
Crybb1 | crystallin, beta B1 | 5 | 2.4 | 1.50E-2 | 173.1 | 407.3 |
Cryba1 | crystallin, beta A1 | 11 | 2.3 | 8.08E-3 | 356.8 | 830.6 |
Crygs | crystallin, gamma S | 16 | 2.3 | 8.58E-3 | 567.8 | 1321.6 |
Lctl | lactase-like | 9 | 2.3 | 2.66E-2 | 7.5 | 17.0 |
Smco3 | single-pass membrane protein with coiled-coil domains 3 | 6 | 2.3 | 5.34E-4 | 2.4 | 5.4 |
Basp1 | brain abundant, membrane attached signal protein 1 | 15 | 2.2 | 8.58E-3 | 39.9 | 87.0 |
Snhg12 | small nucleolar RNA host gene 12 | 4 | 2.2 | 1.06E-2 | 1.8 | 3.9 |
Dpf3 | D4, zinc and double PHD fingers, family 3 | 12 | 2.2 | 8.58E-3 | 1.9 | 4.1 |
Tmem40 | transmembrane protein 40 | 6 | 2.1 | 8.96E-3 | 5.0 | 10.6 |
Arhgap31 | Rho GTPase activating protein 31 | 16 | 2.1 | 5.34E-4 | 2.1 | 4.4 |
Cryba2 | crystallin, beta A2 | 1 | 2.1 | 4.99E-3 | 661.3 | 1371.1 |
Efemp1 | epidermal growth factor-containing fibulin-like extracellular matrix protein 1 | 11 | −2.0 | 2.36E-2 | 33.6 | 16.7 |
Slc6a6 | solute carrier family 6 (neurotransmitter transporter, taurine), member 6 | 6 | −2.0 | 1.83E-4 | 128.6 | 62.7 |
Ntrk2 | neurotrophic tyrosine kinase, receptor, type 2 | 13 | −2.3 | 9.69E-3 | 4.6 | 2.0 |
F5 | coagulation factor V | 1 | −2.3 | 9.80E-3 | 15.2 | 6.5 |
Gpx3 | glutathione peroxidase 3 | 11 | −2.4 | 9.69E-5 | 229.3 | 97.4 |
Matn2 | matrilin 2 | 15 | −2.6 | 3.26E-2 | 5.7 | 2.2 |
Plin4 | perilipin 4 | 17 | −3.2 | 1.76E-2 | 3.7 | 1.2 |
Col9a1 | collagen, type IX, alpha 1 | 1 | −3.5 | 2.00E-6 | 11.9 | 3.4 |
Crhbp | corticotropin releasing hormone binding protein | 13 | −3.5 | 1.18E-3 | 21.4 | 6.2 |
Fbn1 | fibrillin 1 | 2 | −3.7 | 3.58E-10 | 32.3 | 8.6 |
Krt12 | keratin 12 | 11 | −27.8 | 1.18E-3 | 3.5 | 0.1 |
Eif2s3y | eukaryotic translation initiation factor 2, subunit 3, Y-linked | Y | −592.9 | 5.01E-21 | 2.6 | 0.0 |
Ddx3y | DEAD (Asp-Glu-Ala-Asp) box polypeptide 3, Y-linked | Y | −856.2 | 5.65E-93 | 7.8 | 0.0 |
Analysis of these DEGs using iSyte (Kakrana et al., 2018) revealed that 14 DEGs exhibited lens preferred expression in adult lenses. Comparison of genes differentially expressed in male and female aged LECs with those found to differentially expressed between young versus old LECs when sex was not considered as a variable (Faranda et al 2021), revealed that 10 of the 37 genes differentially expressed by sex in aged LECs also significantly change expression level during aging. While the direction of change in males and females was the same in all cases, the extent of these changes was sexually dimorphic (Table 5).
Table 5.
The genes whose expression changes significantly with age in both sexes, and are differentially expressed between lens epithelial cells from aged female mice and male mice.
SYMBOL | DESCRIPTION | aged vs young FC | aged vs young FDR | young FPKM | aged FPKM | aged female vs male FC | aged female vs male FDR | Male aged FPKM | Female aged FPKM |
---|---|---|---|---|---|---|---|---|---|
Cryba1 | crystallin, beta A1 | −3.05 | 3.33E-05 | 1807.54 | 590.89 | 2.33 | 8.08E-03 | 356.79 | 830.56 |
Crybb1 | crystallin, beta B1 | −2.66 | 1.08E-03 | 769.04 | 288.84 | 2.35 | 1.50E-02 | 173.11 | 407.3 |
Slc6a6 | solute carrier family 6 (neurotransmitter transporter, taurine), member 6 | 2.17 | 1.33E-03 | 43.78 | 95.29 | −2.06 | 1.83E-04 | 128.59 | 62.73 |
Crygs | crystallin, gamma S | −2.39 | 3.03E-03 | 2255.19 | 940.23 | 2.33 | 8.58E-03 | 567.75 | 1321.57 |
Fabp5 | fatty acid binding protein 5, epidermal | −3.32 | 4.14E-03 | 104.03 | 31.26 | 3.25 | 1.18E-02 | 14.75 | 48.07 |
Cryba2 | crystallin, beta A2 | −2.01 | 6.83E-03 | 2030.8 | 1011.47 | 2.07 | 4.99E-03 | 661.25 | 1371.15 |
Crygd | crystallin, gamma D | −85.04 | 1.16E-02 | 119.19 | 1.4 | 60.13 | 2.36E-02 | 0.04 | 2.77 |
Cryba4 | crystallin, beta A4 | −2.87 | 1.29E-02 | 1087.55 | 378.32 | 3.25 | 5.90E-03 | 178.99 | 581.28 |
Gpx3 | glutathione peroxidase 3 | 2.66 | 4.68E-02 | 61.17 | 162.73 | −2.36 | 9.69E-05 | 229.28 | 97.36 |
Matn2 | matrilin 2 | 2.31 | 4.93E-02 | 1.71 | 3.95 | −2.62 | 3.26E-02 | 5.75 | 2.19 |
The effect of sex on the response of aged lens epithelial cells to lens fiber cell removal
Comparison between the transcriptome of aged male and female mouse LECs at 24 hours PCS revealed 430 biologically significant DEGs between the sexes, with 267 genes expressed at higher levels in females and 163 expressed at higher levels males. Of these, 17 DEGs reside on sex chromosomes, 8 are encoded on the mitochondrial genome, while the remainder are autosomal (Table 6). Six DEGs located on sex chromosomes, one on the mitochondrial genome, and 90 autosomal DEGs were previously found to exhibit sex dependent expression in other mouse tissues based on comparisons with SAGD (Table 6). Comparison of this list with the iSyTE database revealed that 30 DEGs between male and female LECs at 24 hours PCS exhibit lens preferred expression in adult lenses.
Table 6.
DEGs between aged male and female lens epithelial cells at 24 hours PCS Bold denotes genes that have been previously reported to exhibit sex dependent expression in adult mouse tissues based on comparisons with SAGD (Shi et al., 2019).. Italics denotes genes that exhibit lens preferred expression in postnatal day 56 mouse lenses according to the iSyTe database (Kakrana et al., 2018).
SYMBOL | DESCRIPTION | Chromosome | Fold Change | FDR | Male FPKM | Female FPKM |
---|---|---|---|---|---|---|
Endog | endonuclease G | 2 | 400.3 | 1.52E-5 | 0.0 | 2.7 |
A730094K22Rik | RIKEN cDNA A730094K22 gene | 9 | 361.3 | 2.06E-4 | 0.0 | 3.5 |
Gm24644 | predicted gene, 24644 | 2 | 337.4 | 8.11E-4 | 0.0 | 17.5 |
Gm18840 | predicted gene, 18840 | 6 | 264.2 | 2.65E-2 | 0.0 | 3.7 |
Xist | inactive X specific transcripts | X | 107.5 | 1.47E-2 | 0.5 | 54.8 |
Lipt2 | lipoyl(octanoyl) transferase 2 (putative) | 7 | 21.3 | 2.66E-4 | 0.1 | 2.5 |
Rpl17-ps8 | ribosomal protein L17, pseudogene 8 | X | 13.8 | 3.63E-3 | 0.4 | 5.6 |
Steap4 | STEAP family member 4 | 5 | 13.8 | 3.13E-2 | 2.3 | 31.4 |
Efemp1 | epidermal growth factor-containing fibulin-like extracellular matrix protein 1 | 11 | 13.4 | 1.35E-8 | 4.2 | 57.2 |
Foxi3 | forkhead box I3 | 6 | 12.1 | 2.48E-3 | 0.4 | 4.4 |
Kcnj13 | potassium inwardly-rectifying channel, subfamily J, member 13 | 1 | 12.1 | 9.93E-3 | 0.3 | 3.8 |
Lgi2 | leucine-rich repeat LGI family, member 2 | 5 | 11.3 | 5.49E-4 | 0.5 | 5.5 |
Myl6b | myosin, light polypeptide 6B] | 10 | 11.1 | 4.34E-3 | 0.3 | 3.0 |
C1ra | complement component 1, r subcomponent A | 6 | 10.8 | 2.99E-5 | 0.6 | 6.5 |
Slc6a13 | solute carrier family 6 (neurotransmitter transporter, GABA), member 13 | 6 | 10.4 | 3.98E-4 | 0.6 | 6.8 |
Rida | reactive intermediate imine deaminase A homolog | 15 | 10.3 | 1.71E-3 | 0.3 | 3.5 |
Chil1 | chitinase-like 1 | 1 | 9.9 | 2.76E-2 | 7.9 | 78.4 |
Dio3 | deiodinase, iodothyronine type III | 12 | 9.8 | 2.50E-2 | 1.0 | 10.1 |
Klhdc7a | kelch domain containing 7A | 4 | 9.6 | 2.10E-6 | 0.3 | 3.2 |
Npvf | neuropeptide VF precursor | 6 | 9.5 | 1.33E-2 | 1.3 | 12.2 |
Six6 | sine oculis-related homeobox 6 | 12 | 9.5 | 2.33E-4 | 0.4 | 3.6 |
Tyrp1 | tyrosinase-related protein 1 | 4 | 9.1 | 4.52E-4 | 0.4 | 4.1 |
Mfrp | membrane frizzled-related protein | 9 | 8.9 | 1.06E-3 | 0.5 | 4.6 |
A2m | alpha-2-macroglobulin | 6 | 8.8 | 3.65E-3 | 1.9 | 16.5 |
Bmp4 | bone morphogenetic protein 4 | 14 | 8.7 | 1.52E-3 | 0.7 | 6.5 |
Chrm1 | cholinergic receptor, muscarinic 1, CNS | 19 | 8.7 | 2.39E-4 | 0.4 | 3.5 |
Slc7a4 | solute carrier family 7 (cationic amino acid transporter, y+ system), member 4 | 16 | 8.6 | 5.18E-3 | 0.5 | 4.0 |
Sod3 | superoxide dismutase 3, extracellular | 5 | 8.5 | 4.27E-2 | 18.5 | 156.7 |
Slc16a2 | solute carrier family 16 (monocarboxylic acid transporters), 2 | X | 8.5 | 1.25E-2 | 2.1 | 18.1 |
Rab3b | RAB3B, member RAS oncogene family | 4 | 7.8 | 2.41E-2 | 0.4 | 3.3 |
Rtl8c | retrotransposon Gag like 8C | X | 7.8 | 1.67E-2 | 0.9 | 7.5 |
Macrod1 | mono-ADP ribosylhydrolase 1 | 19 | 7.8 | 1.90E-2 | 0.3 | 2.3 |
Cdh11 | cadherin 11 | 8 | 7.7 | 7.28E-4 | 0.3 | 2.5 |
Mme | membrane metallo endopeptidase | 3 | 7.5 | 3.56E-2 | 1.7 | 13.0 |
Prkcq | protein kinase C, theta | 2 | 7.4 | 5.13E-3 | 0.5 | 4.0 |
Wfdc1 | WAP four-disulfide core domain 1 | 8 | 7.2 | 4.73E-3 | 0.8 | 5.9 |
Otx1 | orthodenticle homeobox 1 | 11 | 7.0 | 4.85E-2 | 0.5 | 3.4 |
Sord | sorbitol dehydrogenase | 2 | 7.0 | 1.49E-2 | 1.0 | 6.8 |
Cldn2 | claudin 2 | X | 6.9 | 6.83E-4 | 0.6 | 4.1 |
Ajap1 | adherens junction associated protein 1 | 4 | 6.4 | 1.14E-2 | 0.4 | 2.8 |
Cavin2 | caveolae associated 2 | 1 | 6.4 | 9.88E-3 | 2.8 | 17.8 |
Map3k21 | mitogen-activated protein kinase kinase kinase 21 | 8 | 6.4 | 7.27E-3 | 0.6 | 3.9 |
Zmat5 | zinc finger, matrin type 5 | 11 | 6.3 | 1.33E-2 | 0.6 | 3.6 |
Rab3il1 | RAB3A interacting protein (rabin3)-like 1 | 19 | 6.3 | 2.11E-2 | 0.4 | 2.5 |
Zic1 | zinc finger protein of the cerebellum 1 | 9 | 6.2 | 2.35E-3 | 1.2 | 7.2 |
Gm14322 | predicted gene 14322 | 2 | 6.2 | 9.97E-3 | 0.5 | 3.2 |
Ephb2 | Eph receptor B2 | 4 | 6.1 | 2.02E-5 | 0.4 | 2.4 |
Selenop | selenoprotein P | 15 | 6.0 | 2.10E-2 | 1.6 | 9.9 |
Pdgfra | platelet derived growth factor receptor, alpha polypeptide | 5 | 6.0 | 4.97E-2 | 0.4 | 2.5 |
Slc13a4 | solute carrier family 13 (sodium/sulfate symporters), member 4 | 6 | 5.8 | 9.49E-4 | 1.5 | 8.9 |
Apcdd1 | adenomatosis polyposis coli down-regulated 1 | 18 | 5.8 | 9.60E-4 | 0.5 | 2.8 |
Serping1 | serine (or cysteine) peptidase inhibitor, clade G, member 1 | 2 | 5.7 | 1.13E-12 | 4.8 | 27.6 |
Aass | aminoadipate-semialdehyde synthase | 6 | 5.6 | 1.62E-2 | 0.5 | 2.9 |
Mrgprf | MAS-related GPR, member F | 7 | 5.6 | 1.64E-2 | 0.5 | 2.9 |
Zic2 | zinc finger protein of the cerebellum 2 | 14 | 5.6 | 2.19E-4 | 1.9 | 10.7 |
Mab21l2 | mab-21-like 2 | 3 | 5.5 | 6.22E-6 | 1.9 | 10.6 |
Lrp2 | low density lipoprotein receptor-related protein 2 | 2 | 5.5 | 6.15E-4 | 1.0 | 5.3 |
Nr2f1 | nuclear receptor subfamily 2, group F, member 1 | 13 | 5.4 | 3.42E-2 | 0.9 | 5.1 |
Cp | ceruloplasmin | 3 | 5.4 | 3.08E-6 | 21.7 | 117.4 |
Tmem176b | transmembrane protein 176B | 6 | 5.4 | 1.24E-7 | 7.0 | 37.9 |
Adamts5 | a disintegrin-like and metallopeptidase (reprolysin type) with thrombospondin type 1 motif, 5 (aggrecanase-2) | 16 | 5.3 | 5.58E-10 | 1.2 | 6.2 |
C1qb | complement component 1, q subcomponent, beta polypeptide | 4 | 5.3 | 9.88E-3 | 1.5 | 8.3 |
Gm14325 | predicted gene 14325 | 2 | 5.1 | 2.51E-2 | 0.8 | 4.4 |
Lsr | lipolysis stimulated lipoprotein receptor | 7 | 5.1 | 4.12E-2 | 0.8 | 4.0 |
F5 | coagulation factor V | 1 | 5.0 | 4.02E-5 | 2.8 | 14.3 |
Bcam | basal cell adhesion molecule | 7 | 5.0 | 1.27E-4 | 1.4 | 7.2 |
Nid2 | nidogen 2 | 14 | 4.8 | 1.20E-2 | 1.9 | 9.1 |
Snapc5 | small nuclear RNA activating complex, polypeptide 5 | 9 | 4.8 | 2.02E-2 | 0.9 | 4.4 |
Rhoj | ras homolog family member J | 12 | 4.6 | 1.12E-2 | 2.7 | 12.4 |
Reln | reelin | 5 | 4.6 | 2.88E-6 | 4.0 | 18.6 |
Fmod | fibromodulin | 1 | 4.6 | 1.89E-5 | 3.7 | 17.0 |
Colgalt2 | collagen beta(1-O)galactosyltransferase 2 | 1 | 4.6 | 3.08E-6 | 1.1 | 5.1 |
Pcyt1b | phosphate cytidylyltransferase 1, choline, beta isoform | X | 4.6 | 2.41E-3 | 1.9 | 8.6 |
Olfml2a | olfactomedin-like 2A | 2 | 4.6 | 2.62E-7 | 4.7 | 21.7 |
Vat1l | vesicle amine transport protein 1 like | 8 | 4.6 | 3.45E-4 | 1.0 | 4.7 |
Hopx | HOP homeobox | 5 | 4.6 | 6.42E-3 | 1.4 | 6.6 |
S1pr3 | sphingosine-1-phosphate receptor 3 | 13 | 4.5 | 4.38E-9 | 6.6 | 29.6 |
Jam2 | junction adhesion molecule 2 | 16 | 4.3 | 1.65E-2 | 1.6 | 6.8 |
Tmem176a | transmembrane protein 176 A | 6 | 4.3 | 2.79E-4 | 2.6 | 11.4 |
Aqp4 | aquaporin 4 | 18 | 4.3 | 8.57E-8 | 3.2 | 13.7 |
Rtn4rl1 | reticulon 4 receptor-like 1 | 11 | 4.3 | 1.26E-3 | 1.9 | 8.2 |
Gpm6a | glycoprotein m6a | 8 | 4.3 | 8.36E-4 | 2.0 | 8.6 |
Il1r1 | interleukin 1 receptor, type I | 1 | 4.3 | 3.56E-2 | 1.2 | 5.2 |
Enpp5 | ectonucleotide pyrophosphatase/phosphodiesterase 5 | 17 | 4.2 | 1.89E-3 | 1.4 | 6.1 |
Trp53i11 | transformation related protein 53 inducible protein 11 | 2 | 4.2 | 2.72E-3 | 1.0 | 4.3 |
Sema3d | sema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3D | 5 | 4.2 | 2.67E-2 | 1.4 | 5.7 |
Fbn1 | fibrillin 1 | 2 | 4.2 | 2.04E-3 | 8.1 | 33.8 |
C1s1 | complement component 1, s subcomponent 1 | 6 | 4.2 | 2.43E-2 | 1.3 | 5.3 |
Mturn | maturin, neural progenitor differentiation regulator homolog | 6 | 4.1 | 2.20E-2 | 1.9 | 7.8 |
Ogn | osteoglycin | 13 | 4.1 | 2.92E-2 | 3.7 | 15.2 |
Klf15 | Kruppel-like factor 15 | 6 | 4.1 | 1.74E-2 | 0.9 | 3.6 |
Rarres2 | retinoic acid receptor responder (tazarotene induced) 2 | 6 | 4.1 | 4.42E-4 | 1.4 | 5.6 |
Napepld | N-acyl phosphatidylethanolamine phospholipase D | 5 | 4.0 | 4.10E-2 | 1.4 | 5.4 |
Cdh4 | cadherin 4 | 2 | 4.0 | 9.60E-4 | 1.5 | 6.0 |
Lrrc75b | leucine rich repeat containing 75B | 10 | 4.0 | 1.90E-2 | 1.0 | 3.9 |
Lypd6 | LY 6/PLAUR domain containing 6 | 2 | 4.0 | 4.40E-2 | 1.2 | 4.7 |
Flrt1 | fibronectin leucine rich transmembrane protein 1 | 19 | 4.0 | 8.28E-5 | 1.5 | 5.8 |
Pcx | pyruvate carboxylase | 19 | 3.9 | 4.63E-5 | 1.0 | 4.0 |
Gpx3 | glutathione peroxidase 3 | 11 | 3.9 | 3.04E-3 | 58.8 | 230.7 |
Zfp982 | zinc finger protein 982 | 4 | 3.9 | 1.02E-2 | 1.2 | 4.6 |
Col18a1 | collagen, type XVIII, alpha 1 | 10 | 3.9 | 3.32E-2 | 23.3 | 91.0 |
Serpinb9 | serine (or cysteine) peptidase inhibitor, clade B, member 9 | 13 | 3.9 | 4.39E-2 | 0.8 | 3.3 |
C4b | complement component 4B (Chido blood group) | 17 | 3.8 | 2.46E-4 | 3.7 | 14.3 |
Amph | amphiphysin | 13 | 3.8 | 1.50E-5 | 5.3 | 20.2 |
Mrc2 | mannose receptor, C type 2 | 11 | 3.8 | 2.70E-2 | 1.2 | 4.6 |
Alad | aminolevulinate, delta-, dehydratase | 4 | 3.8 | 3.07E-2 | 7.3 | 27.7 |
Col1a2 | collagen, type I, alpha 2 | 6 | 3.8 | 9.39E-3 | 0.9 | 3.5 |
Prelp | proline arginine-rich end leucine-rich repeat | 1 | 3.8 | 2.63E-3 | 5.2 | 19.8 |
Optc | opticin | 1 | 3.8 | 7.73E-5 | 21.5 | 81.0 |
Otulinl | OTU deubiquitinase with linear linkage specificity like | 15 | 3.7 | 4.30E-2 | 0.8 | 3.0 |
Sorcs1 | sortilin-related VPS10 domain containing receptor 1 | 19 | 3.7 | 2.34E-2 | 0.8 | 2.9 |
Ephx2 | epoxide hydrolase 2, cytoplasmic | 14 | 3.7 | 1.01E-3 | 3.0 | 10.9 |
Csad | cysteine sulfinic acid decarboxylase | 15 | 3.6 | 4.33E-2 | 3.9 | 13.9 |
Ntrk2 | neurotrophic tyrosine kinase, receptor, type 2 | 13 | 3.6 | 6.51E-3 | 1.0 | 3.4 |
Fbln1 | fibulin 1 | 15 | 3.5 | 3.09E-4 | 15.3 | 54.3 |
Smarcd3 | SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily d, member 3 | 5 | 3.5 | 2.97E-6 | 1.8 | 6.5 |
Mtus1 | mitochondrial tumor suppressor 1 | 8 | 3.5 | 2.89E-4 | 2.0 | 7.0 |
Pla2g5 | phospholipase A2, group V | 4 | 3.5 | 1.71E-3 | 4.7 | 16.5 |
Copz2 | coatomer protein complex, subunit zeta 2 | 11 | 3.5 | 2.54E-2 | 1.3 | 4.4 |
Abat | 4-aminobutyrate aminotransferase | 16 | 3.5 | 9.37E-3 | 2.1 | 7.3 |
Ipp | IAP promoted placental gene | 4 | 3.5 | 4.84E-2 | 0.8 | 2.9 |
Atp2b4 | ATPase, Ca++ transporting, plasma membrane 4 | 1 | 3.5 | 2.18E-4 | 2.0 | 6.9 |
Fundc1 | FUN14 domain containing 1 | X | 3.5 | 1.10E-2 | 3.0 | 10.4 |
Mgst1 | microsomal glutathione S-transferase 1 | 6 | 3.4 | 9.57E-6 | 3.6 | 12.6 |
Ccn5 | cellular communication network factor 5 | 2 | 3.4 | 1.23E-2 | 3.0 | 10.4 |
Hacd4 | 3-hydroxyacyl-CoA dehydratase 4 | 4 | 3.4 | 4.17E-2 | 1.0 | 3.3 |
Galnt12 | polypeptide N-acetylgalactosaminyltransferase 12 | 4 | 3.4 | 3.17E-2 | 1.5 | 5.2 |
Boc | biregional cell adhesion molecule-related/down-regulated by oncogenes (Cdon) binding protein | 16 | 3.4 | 1.84E-3 | 1.6 | 5.4 |
Atp1a2 | ATPase, Na+/K+ transporting, alpha 2 polypeptide | 1 | 3.4 | 6.34E-3 | 29.1 | 99.1 |
Pafah2 | platelet-activating factor acetylhydrolase 2 | 4 | 3.4 | 2.93E-3 | 1.2 | 4.2 |
Cdon | cell adhesion molecule-related/down-regulated by oncogenes | 9 | 3.4 | 2.66E-5 | 4.3 | 14.7 |
Crhbp | corticotropin releasing hormone binding protein | 13 | 3.4 | 4.54E-2 | 5.7 | 19.4 |
Lhx2 | LIM homeobox protein 2 | 2 | 3.4 | 2.22E-2 | 1.6 | 5.3 |
Oplah | 5-oxoprolinase (ATP-hydrolysing) | 15 | 3.3 | 7.22E-4 | 1.5 | 4.9 |
St8sia1 | ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 1 | 6 | 3.3 | 5.52E-3 | 1.2 | 4.1 |
Fcgrt | Fc receptor, IgG, alpha chain transporter | 7 | 3.3 | 3.65E-3 | 2.3 | 7.5 |
Oat | ornithine aminotransferase | 7 | 3.3 | 1.15E-2 | 15.7 | 52.0 |
Vcan | versican | 13 | 3.3 | 7.61E-3 | 4.0 | 13.3 |
Pdk2 | pyruvate dehydrogenase kinase, isoenzyme 2 | 11 | 3.3 | 9.84E-3 | 4.2 | 13.7 |
Col23a1 | collagen, type XXIII, alpha 1 | 11 | 3.3 | 3.96E-2 | 8.1 | 26.7 |
Anapc13 | anaphase promoting complex subunit 13 | 9 | 3.3 | 4.80E-2 | 2.3 | 7.5 |
Scx | scleraxis | 15 | 3.2 | 1.45E-3 | 1.9 | 6.2 |
Cyp39a1 | cytochrome P450, family 39, subfamily a, polypeptide 1 | 17 | 3.2 | 1.19E-2 | 1.2 | 3.9 |
Pdcd4 | programmed cell death 4 | 19 | 3.2 | 2.11E-2 | 17.1 | 54.7 |
Paxx | non-homologous end joining factor | 2 | 3.2 | 2.74E-2 | 2.0 | 6.3 |
Pcolce | procollagen C-endopeptidase enhancer protein | 5 | 3.2 | 5.69E-4 | 3.9 | 12.2 |
Ildr2 | immunoglobulin-like domain containing receptor 2 | 1 | 3.2 | 6.81E-7 | 5.6 | 17.5 |
Gm7694 | predicted gene 7694 | 1 | 3.2 | 3.27E-4 | 3.3 | 10.4 |
Mrpl34 | mitochondrial ribosomal protein L34 | 8 | 3.2 | 4.23E-2 | 3.5 | 11.3 |
Jup | junction plakoglobin | 11 | 3.2 | 1.62E-3 | 1.5 | 4.6 |
Gas1 | growth arrest specific 1 | 13 | 3.2 | 7.41E-6 | 7.6 | 24.0 |
9130023H24Rik | RIKEN cDNA 9130023H24 gene | 7 | 3.1 | 1.44E-2 | 1.0 | 3.1 |
Zfp185 | zinc finger protein 185 | X | 3.1 | 4.76E-5 | 3.3 | 10.2 |
Oxct1 | 3-oxoacid CoA transferase 1 | 15 | 3.1 | 1.03E-4 | 3.1 | 9.5 |
Slc1a3 | solute carrier family 1 (glial high affinity glutamate transporter), 3 | 15 | 3.1 | 5.18E-3 | 1.2 | 3.7 |
Prxl2a | peroxiredoxin like 2A | 14 | 3.1 | 7.30E-6 | 7.4 | 22.7 |
Rcan2 | regulator of calcineurin 2 | 17 | 3.1 | 1.02E-2 | 3.3 | 10.2 |
Isyna1 | myo-inositol 1-phosphate synthase A1 | 8 | 3.1 | 9.03E-3 | 6.2 | 19.0 |
Epm2a | epilepsy, progressive myoclonic epilepsy, type 2 gene alpha | 10 | 3.1 | 2.37E-2 | 1.0 | 3.2 |
Atp1b3 | ATPase, Na+/K+ transporting, beta 3 polypeptide | 9 | 3.0 | 2.43E-2 | 53.6 | 163.6 |
Pnmal2 | PNMA-like 2 | 7 | 3.0 | 1.86E-2 | 5.5 | 16.6 |
Rorc | RAR-related orphan receptor gamma | 3 | 3.0 | 1.91E-2 | 1.7 | 5.1 |
Epas1 | endothelial PAS domain protein 1 | 17 | 3.0 | 1.90E-2 | 5.5 | 16.6 |
Ndufb4 | NADH:ubiquinone oxidoreductase subunit B4 | 16 | 3.0 | 8.21E-3 | 4.2 | 12.6 |
Ech1 | enoyl coenzyme A hydratase 1, peroxisomal | 7 | 3.0 | 7.41E-4 | 2.4 | 7.0 |
Ndrg1 | N-myc downstream regulated gene 1 | 15 | 3.0 | 7.59E-3 | 8.0 | 23.7 |
Acadsb | acyl-Coenzyme A dehydrogenase, short/branched chain | 7 | 3.0 | 1.52E-3 | 2.4 | 7.1 |
Scara3 | scavenger receptor class A, member 3 | 14 | 3.0 | 1.02E-2 | 1.5 | 4.4 |
Lix1l | Lix1-like | 3 | 3.0 | 1.34E-4 | 3.2 | 9.3 |
Abi3bp | ABI gene family, member 3 (NESH) binding protein | 16 | 2.9 | 3.12E-3 | 4.4 | 12.9 |
Stra6 | stimulated by retinoic acid gene 6 | 9 | 2.9 | 2.66E-2 | 1.3 | 4.0 |
Sntb1 | syntrophin, basic 1 | 15 | 2.9 | 9.84E-3 | 3.3 | 9.7 |
Ephb6 | Eph receptor B6 | 6 | 2.9 | 7.79E-3 | 1.3 | 3.7 |
Loxl1 | lysyl oxidase-like 1 | 9 | 2.9 | 2.29E-5 | 11.3 | 33.1 |
Fat4 | FAT atypical cadherin 4 | 3 | 2.9 | 3.62E-3 | 6.1 | 17.9 |
Itgb8 | integrin beta 8 | 12 | 2.9 | 2.06E-2 | 1.8 | 5.2 |
Sesn1 | sestrin 1 | 10 | 2.9 | 1.94E-2 | 3.3 | 9.6 |
Irs1 | insulin receptor substrate 1 | 1 | 2.9 | 3.46E-3 | 2.0 | 5.8 |
Gldc | glycine decarboxylase | 19 | 2.9 | 1.62E-3 | 5.0 | 14.3 |
Porcn | porcupine O-acyltransferase | X | 2.9 | 1.12E-3 | 3.4 | 9.7 |
Gulp1 | GULP, engulfment adaptor PTB domain containing 1 | 1 | 2.9 | 1.52E-2 | 1.3 | 3.8 |
Sqor | sulfide quinone oxidoreductase | 2 | 2.9 | 3.69E-3 | 1.1 | 3.1 |
Fbxo10 | F-box protein 10 | 4 | 2.9 | 1.23E-3 | 1.4 | 4.1 |
Dll 1 | delta like canonical Notch ligand 1 | 17 | 2.9 | 1.92E-2 | 2.4 | 6.7 |
Plaat3 | phospholipase A and acyltransferase 3 | 19 | 2.9 | 3.56E-2 | 1.8 | 5.1 |
Ptgds | prostaglandin D2 synthase (brain) | 2 | 2.8 | 1.37E-3 | 116.1 | 329.2 |
Dync2li 1 | dynein cytoplasmic 2 light intermediate chain 1 | 17 | 2.8 | 4.17E-2 | 1.5 | 4.2 |
Ndufa4 | Ndufa4, mitochondrial complex associated | 6 | 2.8 | 4.72E-2 | 2.2 | 6.2 |
Zhx1 | zinc fingers and homeoboxes 1 | 15 | 2.8 | 1.95E-2 | 2.2 | 6.3 |
Alg14 | asparagine-linked glycosylation 14 | 3 | 2.8 | 1.71E-2 | 1.3 | 3.6 |
Chchd10 | coiled-coil-helix-coiled-coil-helix domain containing 10 | 10 | 2.8 | 4.27E-4 | 7.0 | 19.4 |
Fads2 | fatty acid desaturase 2 | 19 | 2.8 | 2.70E-4 | 6.9 | 19.2 |
Surf1 | surfeit gene 1 | 2 | 2.7 | 6.80E-3 | 3.3 | 9.1 |
Pdpr | pyruvate dehydrogenase phosphatase regulatory subunit | 8 | 2.7 | 5.96E-3 | 1.3 | 3.5 |
Rbms2 | RNA binding motif, single stranded interacting protein 2 | 10 | 2.7 | 5.19E-3 | 1.7 | 4.7 |
Idh1 | isocitrate dehydrogenase 1 (NADP+), soluble | 1 | 2.7 | 3.60E-3 | 3.3 | 8.9 |
Rnf43 | ring finger protein 43] | 11 | 2.7 | 7.45E-3 | 1.3 | 3.6 |
Aldh6a1 | aldehyde dehydrogenase family 6, subfamily A1 | 12 | 2.7 | 2.89E-2 | 1.6 | 4.2 |
Parm1 | prostate androgen-regulated mucin-like protein 1 | 5 | 2.6 | 1.26E-2 | 3.4 | 9.1 |
Igfbp4 | insulin-like growth factor binding protein 4 | 11 | 2.6 | 2.02E-2 | 6.9 | 18.2 |
Shisa2 | shisa family member 2 | 14 | 2.6 | 4.95E-2 | 1.6 | 4.2 |
Fam107a | family with sequence similarity 107, member A | 14 | 2.6 | 2.76E-2 | 2.5 | 6.5 |
Hltf | helicase-like transcription factor | 3 | 2.6 | 5.11E-3 | 1.7 | 4.3 |
Gm44250 | predicted gene, 44250 | 6 | 2.6 | 1.54E-2 | 1.3 | 3.3 |
Slc4a5 | solute carrier family 4, sodium bicarbonate cotransporter, member 5 | 6 | 2.6 | 2.86E-2 | 3.0 | 7.7 |
Scp2 | sterol carrier protein 2, liver [ | 4 | 2.6 | 9.03E-3 | 9.4 | 24.1 |
Antxr1 | anthrax toxin receptor 1 | 6 | 2.6 | 5.58E-3 | 5.3 | 13.5 |
Rdh10 | retinol dehydrogenase 10 (all-trans) | 1 | 2.5 | 1.03E-2 | 8.9 | 22.7 |
Slc22a17 | solute carrier family 22 (organic cation transporter), member 17 | 14 | 2.5 | 1.06E-2 | 2.8 | 7.2 |
Apoe | apolipoprotein E | 7 | 2.5 | 1.18E-2 | 238.9 | 605.0 |
Acsl3 | acyl-CoA synthetase long-chain family member 3 | 1 | 2.5 | 7.41E-4 | 6.2 | 15.7 |
Ace | angiotensin I converting enzyme (peptidyl-dipeptidase A) 1 | 11 | 2.5 | 1.03E-2 | 1.5 | 3.9 |
Scd1 | stearoyl-Coenzyme A desaturase 1 | 19 | 2.5 | 4.13E-2 | 3.1 | 7.8 |
Ldhb | lactate dehydrogenase B | 6 | 2.5 | 1.15E-2 | 8.9 | 22.4 |
Crb2 | crumbs family member 2 | 2 | 2.5 | 3.34E-2 | 3.2 | 8.1 |
Scel | sciellin | 14 | 2.5 | 2.03E-2 | 12.3 | 30.8 |
Tsc22d1 | TSC22 domain family, member 1 | 14 | 2.5 | 2.93E-3 | 30.2 | 74.8 |
Sod2 | superoxide dismutase 2, mitochondrial | 17 | 2.5 | 1.61E-3 | 3.0 | 7.4 |
Kcnk1 | potassium channel, subfamily K, member 1 | 8 | 2.4 | 1.18E-3 | 7.4 | 18.2 |
Tmem106c | transmembrane protein 106C | 15 | 2.4 | 1.63E-2 | 3.4 | 8.3 |
Tenm4 | teneurin transmembrane protein 4 | 7 | 2.4 | 1.61E-3 | 3.1 | 7.5 |
Gas6 | growth arrest specific 6 | 8 | 2.4 | 4.05E-3 | 15.5 | 37.7 |
Tns2 | tensin 2 | 15 | 2.4 | 2.76E-2 | 3.9 | 9.4 |
Cox7b | cytochrome c oxidase subunit 7B | X | 2.4 | 1.90E-2 | 11.0 | 26.2 |
Vps41 | VPS41 HOPS complex subunit | 13 | 2.4 | 4.16E-3 | 2.7 | 6.3 |
Ddah2 | dimethylarginine dimethylaminohydrolase 2 | 17 | 2.4 | 1.47E-2 | 5.2 | 12.3 |
Sort1 | sortilin 1 | 3 | 2.4 | 1.25E-3 | 4.5 | 10.5 |
Cd99l2 | CD99 antigen-like 2 | X | 2.4 | 1.13E-2 | 4.6 | 10.9 |
Gpc6 | glypican 6 | 14 | 2.3 | 2.96E-2 | 1.8 | 4.3 |
Rfxap | regulatory factor X-associated protein | 3 | 2.3 | 4.49E-2 | 3.7 | 8.6 |
Ids | iduronate 2-sulfatase | X | 2.3 | 1.57E-2 | 2.8 | 6.4 |
Tspan7 | tetraspanin 7 | X | 2.3 | 7.47E-3 | 2.5 | 5.8 |
Efna5 | ephrin A5 | 17 | 2.3 | 2.69E-3 | 5.0 | 11.6 |
Phactr2 | phosphatase and actin regulator 2 | 10 | 2.3 | 9.39E-3 | 4.1 | 9.3 |
Nfia | nuclear factor I/A | 4 | 2.3 | 8.76E-3 | 2.7 | 6.1 |
Tmem94 | transmembrane protein 94 | 11 | 2.3 | 2.35E-2 | 3.6 | 8.3 |
Yipf4 | Yip1 domain family, member 4 | 17 | 2.3 | 8.43E-3 | 4.3 | 9.8 |
Ube2d-ps | ubiquitin-conjugating enzyme E2D, pseudogene | 11 | 2.3 | 3.94E-2 | 2.1 | 4.8 |
Cxxc5 | CXXC finger 5 | 18 | 2.3 | 2.85E-3 | 5.6 | 12.8 |
Gpc4 | glypican 4 | X | 2.3 | 5.44E-3 | 8.0 | 18.3 |
Mmut | methylmalonyl-Coenzyme A mutase | 17 | 2.3 | 4.60E-2 | 2.3 | 5.3 |
Zfp52 | zinc finger protein 52 | 17 | 2.3 | 3.90E-2 | 2.1 | 4.7 |
Nek7 | NIMA (never in mitosis gene a)-related expressed kinase 7 | 1 | 2.3 | 3.42E-2 | 2.4 | 5.5 |
Ptpn13 | protein tyrosine phosphatase, non-receptor type 13 | 5 | 2.2 | 3.13E-4 | 2.3 | 5.1 |
Cst3 | cystatin C | 2 | 2.2 | 1.02E-2 | 64.4 | 144.5 |
Uqcr11 | ubiquinol-cytochrome c reductase, complex III subunit XI | 10 | 2.2 | 3.00E-2 | 8.7 | 19.5 |
Cadm2 | cell adhesion molecule 2 | 16 | 2.2 | 2.19E-2 | 4.2 | 9.3 |
Phyh | phytanoyl-CoA hydroxylase | 2 | 2.2 | 7.59E-3 | 5.7 | 12.7 |
Insig1 | insulin induced gene i | 5 | 2.2 | 2.55E-2 | 6.7 | 14.8 |
Bcl6 | B cell leukemia/lymphoma 6 | 16 | 2.2 | 3.22E-2 | 2.6 | 5.8 |
Fads1 | fatty acid desaturase 1 | 19 | 2.2 | 9.88E-3 | 9.5 | 21.0 |
Gprc5c | G protein-coupled receptor, family C, group 5, member C | 11 | 2.2 | 3.13E-2 | 3.5 | 7.6 |
Pygb | brain glycogen phosphorylase | 2 | 2.2 | 9.93E-3 | 5.5 | 12.0 |
Gpalpp1 | GPALPP motifs containing 1 | 14 | 2.1 | 3.35E-2 | 2.6 | 5.5 |
Etv5 | ets variant 5 | 16 | 2.1 | 4.17E-2 | 5.9 | 12.5 |
Laptm4a | lysosomal-associated protein transmembrane 4A | 12 | 2.1 | 2.37E-2 | 25.8 | 54.1 |
Il11ra1 | interleukin 11 receptor, alpha chain 1 | 4 | 2.1 | 3.52E-2 | 3.3 | 7.0 |
Dusp3 | dual specificity phosphatase 3 | 11 | 2.1 | 1.35E-2 | 3.1 | 6.4 |
Slc7a11 | solute carrier family 7 (cationic amino acid transporter, y+ system), member 11 | 3 | 2.1 | 1.33E-2 | 13.7 | 28.5 |
Igsf8 | immunoglobulin superfamily, member 8 | 1 | 2.1 | 4.85E-2 | 3.4 | 7.1 |
Scrn1 | secernin 1 | 6 | 2.1 | 4.56E-2 | 4.0 | 8.4 |
Prickle2 | prickle planar cell polarity protein 2 | 6 | 2.1 | 1.23E-2 | 2.4 | 4.9 |
Axin2 | axin 2 | 11 | 2.1 | 2.76E-2 | 3.3 | 6.9 |
Fdft1 | farnesyl diphosphate farnesyl transferase 1 | 14 | 2.1 | 1.02E-2 | 5.0 | 10.3 |
Creg1 | cellular repressor of E1A-stimulated genes 1 | 1 | 2.1 | 3.23E-2 | 5.9 | 12.1 |
Pdhx | pyruvate dehydrogenase complex, component X | 2 | 2.0 | 3.18E-2 | 3.1 | 6.4 |
Ndufb10 | NADH:ubiquinone oxidoreductase subunit B10 | 17 | 2.0 | 4.16E-2 | 16.8 | 34.2 |
Arf6 | ADP-ribosylation factor 6 | 12 | −2.0 | 2.59E-2 | 66.5 | 32.9 |
Pprc1 | peroxisome proliferative activated receptor, gamma, coactivator-related 1 | 19 | −2.0 | 3.52E-2 | 18.2 | 9.0 |
Eif6 | eukaryotic translation initiation factor 6 | 2 | −2.0 | 4.24E-2 | 40.2 | 19.8 |
Sema4d | sema domain, immunoglobulin domain (Ig), transmembrane domain(TM) and short cytoplasmic domain, (semaphorin) 4D | 13 | −2.0 | 6.13E-3 | 10.6 | 5.2 |
Noc2l | NOC2 like nucleolar associated transcriptional repressor | 4 | −2.0 | 3.11E-2 | 47.6 | 23.4 |
Bcar1 | breast cancer anti-estrogen resistance 1 | 8 | −2.1 | 3.42E-2 | 33.2 | 16.2 |
Bach2 | BTB and CNC homology, basic leucine zipper transcription factor 2 | 4 | −2.1 | 1.66E-2 | 4.8 | 2.3 |
Ero1l | ERO1-like (S. cerevisiae) | 14 | −2.1 | 1.47E-2 | 8.6 | 4.2 |
Cttnbp2nl | CTTNBP2 N-terminal like | 3 | −2.1 | 4.67E-2 | 16.7 | 8.0 |
Abl2 | v-abl Abelson murine leukemia viral oncogene 2 (arg, Abelson-related gene) | 1 | −2.1 | 8.19E-3 | 10.5 | 5.0 |
Trim25 | tripartite motif-containing 25 | 11 | −2.1 | 1.79E-2 | 14.3 | 6.8 |
Rapgef3 | Rap guanine nucleotide exchange factor (GEF) 3 | 15 | −2.1 | 6.66E-3 | 14.1 | 6.7 |
Card10 | caspase recruitment domain family, member 10 | 15 | −2.1 | 4.40E-2 | 17.6 | 8.4 |
Ccdc137 | coiled-coil domain containing 137 | 11 | −2.1 | 3.74E-2 | 8.1 | 3.8 |
Pttg 1ip | pituitary tumor-transforming 1 interacting protein | 10 | −2.1 | 4.69E-2 | 26.7 | 12.6 |
Lrrfip2 | leucine rich repeat (in FLII) interacting protein 2 | 9 | −2.2 | 2.60E-2 | 26.0 | 12.1 |
Prtg | protogenin | 9 | −2.2 | 1.35E-2 | 4.3 | 2.0 |
Tjap1 | tight junction associated protein 1 | 17 | −2.2 | 1.48E-2 | 12.5 | 5.8 |
Ppp1r15a | protein phosphatase 1, regulatory subunit 15A | 7 | −2.2 | 9.93E-3 | 28.1 | 12.9 |
Ehd1 | EH-domain containing 1 | 19 | −2.2 | 1.57E-2 | 58.3 | 26.7 |
Vasp | vasodilator-stimulated phosphoprotein | 7 | −2.2 | 4.84E-2 | 60.0 | 27.5 |
Erc2 | ELKS/RAB6-interacting/CAST family member 2 | 14 | −2.2 | 4.80E-2 | 8.3 | 3.8 |
Shb | src homology 2 domain-containing transforming protein B | 4 | −2.2 | 1.02E-2 | 21.0 | 9.6 |
Vps37b | vacuolar protein sorting 37B | 5 | −2.2 | 1.69E-2 | 15.1 | 6.9 |
Rnf217 | ring finger protein 217 | 10 | −2.2 | 2.27E-2 | 5.0 | 2.3 |
Sart3 | squamous cell carcinoma antigen recognized by T cells 3 | 5 | −2.2 | 5.32E-3 | 20.4 | 9.2 |
Eaf1 | ELL associated factor 1 | 14 | −2.2 | 3.56E-2 | 20.2 | 9.1 |
Ccdc9b | coiled-coil domain containing 9B | 2 | −2.2 | 8.44E-3 | 33.4 | 15.1 |
Rhbdf2 | rhomboid 5 homolog 2 | 11 | −2.2 | 1.18E-2 | 12.0 | 5.4 |
Ftsj3 | FtsJ RNA methyltransferase homolog 3 (E. coli) | 11 | −2.2 | 9.26E-3 | 24.7 | 11.1 |
Cdc42ep3 | CDC42 effector protein (Rho GTPase binding) 3 | 17 | −2.2 | 4.99E-2 | 8.7 | 3.9 |
Ctu2 | cytosolic thiouridylase subunit 2 | 8 | −2.3 | 4.81E-2 | 5.1 | 2.3 |
Litaf | LPS-induced TN factor | 16 | −2.3 | 1.44E-2 | 15.0 | 6.6 |
Surf6 | surfeit gene 6 | 2 | −2.3 | 1.05E-3 | 13.9 | 6.1 |
Phldb1 | pleckstrin homology like domain, family B, member 1 | 9 | −2.3 | 2.42E-3 | 44.3 | 19.4 |
Igsf9b | immunoglobulin superfamily, member 9B | 9 | −2.3 | 4.40E-2 | 4.9 | 2.1 |
Klhdc4 | kelch domain containing 4 | 8 | −2.3 | 2.33E-3 | 18.1 | 7.9 |
Tatdn2 | TatD DNase domain containing 2 | 6 | −2.3 | 1.60E-3 | 41.9 | 18.3 |
mt-Nd2 | mitochondrially encoded NADH dehydrogenase 2 | MT | −2.3 | 4.41E-2 | 3117.5 | 1361.6 |
Nol10 | nucleolar protein 10 | 12 | −2.3 | 1.96E-2 | 22.2 | 9.6 |
Srf | serum response factor | 17 | −2.3 | 1.06E-2 | 32.5 | 14.0 |
Urb2 | URB2 ribosome biogenesis 2 homolog (S. cerevisiae) | 8 | −2.3 | 3.10E-2 | 5.0 | 2.1 |
Urb1 | URB1 ribosome biogenesis 1 homolog (S. cerevisiae) | 16 | −2.3 | 3.88E-2 | 4.5 | 1.9 |
Smad7 | SMAD family member 7 | 18 | −2.3 | 1.18E-2 | 33.8 | 14.5 |
Trim47 | tripartite motif-containing 47 | 11 | −2.3 | 1.68E-2 | 24.3 | 10.4 |
Gprc5a | G protein-coupled receptor, family C, group 5, member A | 6 | −2.3 | 3.13E-2 | 7.5 | 3.2 |
Epha2 | Eph receptor A2 | 4 | −2.4 | 3.62E-3 | 42.1 | 17.9 |
Zswim4 | zinc finger SWIM-type containing 4 | 8 | −2.4 | 1.02E-2 | 19.3 | 8.1 |
Mical2 | microtubule associated monooxygenase, calponin and LIM domain containing 2 | 7 | −2.4 | 7.81E-3 | 13.0 | 5.5 |
Tnfaip3 | tumor necrosis factor, alpha-induced protein 3 | 10 | −2.4 | 1.25E-3 | 20.7 | 8.7 |
Sh3bp2 | SH3-domain binding protein 2 | 5 | −2.4 | 4.84E-2 | 8.6 | 3.6 |
Kcnd3 | potassium voltage-gated channel, Shal-related family, member 3 | 3 | −2.4 | 1.30E-2 | 5.3 | 2.2 |
Kif7 | kinesin family member 7 | 7 | −2.4 | 4.84E-2 | 3.9 | 1.6 |
5430416N02Rik | RIKEN cDNA 5430416N02 gene | 5 | −2.4 | 4.17E-2 | 4.8 | 2.0 |
Rnf213 | ring finger protein 213 | 11 | −2.4 | 1.10E-2 | 29.3 | 12.0 |
Rrp9 | RRP9, small subunit (SSU) processome component, homolog (yeast) | 9 | −2.4 | 1.18E-2 | 16.3 | 6.7 |
mt-Nd4 | mitochondrially encoded NADH dehydrogenase 4 | MT | −2.4 | 2.05E-2 | 1988.1 | 815.8 |
mt-Cytb | mitochondrially encoded cytochrome b | MT | −2.4 | 2.34E-2 | 7413.0 | 3027.4 |
Tubb2b | tubulin, beta 2B class IIB | 13 | −2.5 | 2.29E-2 | 18.9 | 7.6 |
Gm24187 | predicted gene, 24187 | 13 | −2.5 | 3.56E-2 | 3979.7 | 1607.5 |
Oasl2 | 2’-5’ oligoadenylate synthetase-like 2 | 5 | −2.5 | 1.70E-3 | 18.5 | 7.5 |
mt-Nd6 | mitochondrially encoded NADH dehydrogenase 6 | MT | −2.5 | 3.10E-2 | 851.8 | 342.4 |
Emilin2 | elastin microfibril interfacer 2 | 17 | −2.5 | 7.19E-3 | 25.2 | 10.0 |
Nbeal2 | neurobeachin-like 2 | 9 | −2.5 | 9.16E-3 | 4.9 | 2.0 |
Plec | plectin | 15 | −2.5 | 4.68E-3 | 466.1 | 184.4 |
Snhg17 | small nucleolar RNA host gene 17 | 2 | −2.5 | 2.43E-2 | 4.0 | 1.6 |
Helz2 | helicase with zinc finger 2, transcriptional coactivator | 2 | −2.5 | 4.29E-2 | 8.6 | 3.4 |
Klf16 | Kruppel-like factor 16 | 10 | −2.5 | 1.68E-2 | 7.9 | 3.1 |
Card19 | caspase recruitment domain family, member 19 | 13 | −2.5 | 3.75E-2 | 36.2 | 14.2 |
mt-Co1 | mitochondrially encoded cytochrome c oxidase I | MT | −2.6 | 2.85E-2 | 6096.8 | 2387.9 |
Tnk2 | tyrosine kinase, non-receptor, 2 | 16 | −2.6 | 1.01E-3 | 25.7 | 9.9 |
Cd274 | CD274 antigen | 19 | −2.6 | 4.85E-2 | 12.2 | 4.7 |
Gm8995 | predicted gene 8995 | 7 | −2.6 | 2.19E-2 | 5.0 | 1.9 |
Ier5 | immediate early response 5 | 1 | −2.7 | 8.19E-3 | 41.0 | 15.5 |
Ccdc88b | coiled-coil domain containing 88B | 19 | −2.7 | 1.68E-2 | 15.2 | 5.7 |
Fmnl1 | formin-like 1 | 11 | −2.7 | 4.57E-2 | 30.6 | 11.5 |
Oaf | out at first homolog | 9 | −2.7 | 9.13E-4 | 40.8 | 15.3 |
Pik3ap1 | phosphoinositide-3-kinase adaptor protein 1 | 19 | −2.7 | 5.32E-3 | 17.2 | 6.4 |
mt-Rnr2 | mitochondrially encoded 16S rRNA | MT | −2.7 | 3.72E-2 | 111335.5 | 41317.1 |
Tcirg1 | T cell, immune regulator 1, ATPase, H+ transporting, lysosomal V0 protein A3 | 19 | −2.7 | 2.05E-2 | 24.3 | 8.9 |
Plk3 | polo like kinase 3 | 4 | −2.7 | 2.94E-2 | 17.3 | 6.3 |
mt-Rnr1 | mitochondrially encoded 12S rRNA | MT | −2.8 | 3.49E-2 | 93896.8 | 33926.5 |
Il1rn | interleukin 1 receptor antagonist | 2 | −2.8 | 3.15E-2 | 31.0 | 11.2 |
Slfn5 | schlafen 5 | 11 | −2.8 | 3.07E-2 | 6.6 | 2.4 |
Mtmr10 | myotubularin related protein 10 | 7 | −2.8 | 2.73E-3 | 15.6 | 5.6 |
Mertk | MER proto-oncogene tyrosine kinase | 2 | −2.8 | 3.04E-3 | 10.2 | 3.7 |
Dot1l | DOT1-like, histone H3 methyltransferase (S. cerevisiae) | 10 | −2.8 | 3.35E-4 | 28.6 | 10.2 |
Smox | spermine oxidase | 2 | −2.8 | 1.09E-2 | 23.3 | 8.3 |
Pim1 | proviral integration site 1 | 17 | −2.8 | 2.04E-2 | 56.7 | 19.9 |
Syk | spleen tyrosine kinase | 13 | −2.8 | 2.03E-2 | 11.0 | 3.9 |
Gm10925 | predicted gene 10925 | 1 | −2.9 | 4.84E-2 | 31.0 | 10.8 |
Rnf19b | ring finger protein 19B | 4 | −2.9 | 3.42E-2 | 52.9 | 18.4 |
Lrrfip1 | leucine rich repeat (in FLII) interacting protein 1 | 1 | −3.0 | 1.16E-3 | 97.8 | 32.4 |
Dusp7 | dual specificity phosphatase 7 | 9 | −3.0 | 2.05E-3 | 38.0 | 12.5 |
Arl4d | ADP-ribosylation factor-like 4D | 11 | −3.0 | 2.33E-3 | 36.9 | 12.1 |
Dimt1 | DIM1 dimethyladenosine transferase 1-like (S. cerevisiae) | 13 | −3.1 | 7.97E-4 | 8.4 | 2.7 |
Gm48958 | predicted gene, 48958 | 14 | −3.1 | 2.86E-2 | 7.7 | 2.5 |
Cmtr2 | cap methyltransferase 2 | 8 | −3.1 | 1.24E-2 | 3.7 | 1.2 |
Nrg1 | neuregulin 1 | 8 | −3.1 | 4.11E-2 | 3.7 | 1.2 |
Fjx1 | four jointed box 1 | 2 | −3.1 | 3.42E-2 | 10.4 | 3.3 |
Thoc6 | THO complex 6 | 17 | −3.1 | 2.70E-2 | 9.9 | 3.2 |
Cdt1 | chromatin licensing and DNA replication factor 1 | 8 | −3.1 | 4.32E-2 | 10.3 | 3.3 |
Slc16a3 | solute carrier family 16 (monocarboxylic acid transporters), 3 | 11 | −3.2 | 1.30E-3 | 15.0 | 4.7 |
Elf3 | E74-like factor 3 [ | 1 | −3.2 | 3.34E-2 | 9.7 | 3.0 |
Acod1 | aconitate decarboxylase 1 | 14 | −3.2 | 1.84E-2 | 24.6 | 7.6 |
Arhgap22 | Rho GTPase activating protein 22 | 14 | −3.3 | 1.34E-4 | 18.0 | 5.5 |
G0s2 | G0/G1 switch gene 2 | 1 | −3.3 | 1.13E-3 | 83.1 | 25.5 |
Csf3r | colony stimulating factor 3 receptor (granulocyte) | 4 | −3.3 | 3.99E-3 | 51.9 | 15.7 |
Baiap2 | brain-specific angiogenesis inhibitor 1-associated protein 2 | 11 | −3.3 | 1.22E-2 | 11.3 | 3.4 |
Sp100 | nuclear antigen Sp100 | 1 | −3.3 | 2.74E-2 | 3.8 | 1.1 |
Sfn | stratifin | 4 | −3.3 | 3.56E-2 | 14.4 | 4.3 |
Atf3 | activating transcription factor 3 | 1 | −3.4 | 3.50E-2 | 31.4 | 9.3 |
Srxn1 | sulfiredoxin 1 homolog (S. cerevisiae) | 2 | −3.4 | 1.32E-2 | 67.9 | 20.0 |
Phlda1 | pleckstrin homology like domain, family A, member 1 | 10 | −3.4 | 3.60E-3 | 77.3 | 22.8 |
Chka | choline kinase alpha | 19 | −3.5 | 1.41E-5 | 15.6 | 4.5 |
Tsc22d2 | TSC22 domain family, member 2 | 3 | −3.5 | 9.57E-6 | 27.5 | 7.9 |
Gm44899 | predicted gene 44899 | 7 | −3.5 | 4.35E-2 | 6.1 | 1.7 |
Ifrd1 | interferon-related developmental regulator 1 | 12 | −3.6 | 1.91E-3 | 22.2 | 6.1 |
Mxd1 | MAX dimerization protein 1 | 6 | −3.7 | 6.51E-3 | 37.5 | 10.2 |
Spata5l1 | spermatogenesis associated 5-like 1 | 2 | −3.7 | 4.45E-2 | 11.3 | 3.1 |
Pxdc1 | PX domain containing 1 | 13 | −3.8 | 4.39E-2 | 7.0 | 1.9 |
Dusp4 | dual specificity phosphatase 4 | 8 | −3.8 | 3.17E-2 | 20.4 | 5.4 |
Nefm | neurofilament, medium polypeptide | 14 | −3.9 | 1.02E-2 | 12.4 | 3.2 |
Maff | v-maf musculoaponeurotic fibrosarcoma oncogene family, protein F | 15 | −4.0 | 5.19E-4 | 20.6 | 5.2 |
H2-Q4 | histocompatibility 2, Q region locus 4 | 17 | −4.1 | 1.32E-2 | 36.4 | 8.9 |
Borcs6 | BLOC-1 related complex subunit 6 | 11 | −4.2 | 2.92E-2 | 4.8 | 1.1 |
Gadd45g | growth arrest and DNA-damage-inducible 45 gamma | 13 | −4.3 | 2.45E-2 | 140.7 | 32.4 |
Kdelr3 | KDEL endoplasmic reticulum protein retention receptor 3 | 15 | −4.4 | 2.42E-3 | 10.0 | 2.3 |
Neat1 | nuclear paraspeckle assembly transcript 1 (non-protein coding) | 19 | −4.4 | 9.13E-4 | 94.8 | 21.7 |
Gm36940 | predicted gene, 36940 | 9 | −4.7 | 4.29E-2 | 3.0 | 0.6 |
Ngf | nerve growth factor | 3 | −4.8 | 3.94E-2 | 42.0 | 8.8 |
Ifi204 | interferon activated gene 204 | 1 | −5.0 | 1.71E-2 | 14.2 | 2.8 |
mt-Atp6 | mitochondrially encoded ATP synthase 6 | MT | −5.0 | 1.23E-2 | 37.7 | 7.5 |
Rn7s2 | 7S RNA 2 | 12 | −5.0 | 3.12E-3 | 101.4 | 20.2 |
Rbp4 | retinol binding protein 4, plasma | 19 | −5.1 | 3.18E-2 | 6.5 | 1.3 |
Flnc | filamin C, gamma | 6 | −5.8 | 3.35E-2 | 178.3 | 30.6 |
Cyp26a1 | cytochrome P450, family 26, subfamily a, polypeptide 1 | 19 | −6.0 | 1.01E-3 | 67.8 | 11.3 |
Mirt2 | myocardial infraction associated transcript 2 | 15 | −6.6 | 3.13E-2 | 4.3 | 0.7 |
Gm23833 | predicted gene, 23833 | 17 | −6.6 | 4.33E-3 | 29.7 | 4.5 |
Trpc6 | transient receptor potential cation channel, subfamily C, member 6 | 9 | −6.6 | 3.61E-3 | 4.3 | 0.7 |
Rsad2 | radical S-adenosyl methionine domain containing 2 | 12 | −7.1 | 2.01E-2 | 22.9 | 3.2 |
Gm26532 | predicted gene, 26532 | 8 | −7.8 | 3.35E-2 | 5.4 | 0.7 |
Tnfsf14 | tumor necrosis factor (ligand) superfamily, member 14 | 17 | −8.7 | 4.19E-2 | 3.2 | 0.4 |
Emp1 | epithelial membrane protein 1 | 6 | −9.2 | 4.95E-2 | 59.6 | 6.5 |
Oasl1 | 2’-5’ oligoadenylate synthetase-like 1 | 5 | −9.7 | 3.62E-3 | 13.1 | 1.3 |
Mir3109 | microRNA 3109 | 9 | −9.9 | 2.92E-2 | 58.0 | 5.8 |
Muc5ac | mucin 5, subtypes A and C, tracheobronchial/gastric | 7 | −10.2 | 2.97E-6 | 5.4 | 0.5 |
Cfap58 | cilia and flagella associated protein 58 | 19 | −11.3 | 6.99E-3 | 3.4 | 0.3 |
Foxn1 | forkhead box N1 | 11 | −11.7 | 2.93E-3 | 3.5 | 0.3 |
Asprv1 | aspartic peptidase, retroviral-like 1 | 6 | −13.9 | 6.66E-3 | 14.8 | 1.1 |
Klk9 | kallikrein related-peptidase 9 | 7 | −14.0 | 2.11E-2 | 3.7 | 0.3 |
Gm44275 | predicted gene, 44275 | 6 | −14.3 | 4.44E-2 | 3.2 | 0.2 |
Krt16 | keratin 16 | 11 | −16.8 | 4.29E-2 | 16.8 | 1.0 |
Krt17 | keratin 17 | 11 | −23.4 | 2.33E-3 | 18.3 | 0.8 |
Muc4 | mucin 4 | 16 | −25.4 | 1.74E-2 | 4.4 | 0.2 |
Gm9573 | predicted gene 9573 | 17 | −45.5 | 3.47E-2 | 3.1 | 0.1 |
Irx1 | Iroquois homeobox 1 | 13 | −69.9 | 2.76E-3 | 2.2 | 0.0 |
Krt12 | keratin 12 | 11 | −85.9 | 1.88E-4 | 6.9 | 0.1 |
C2cd4b | C2 calcium-dependent domain containing 4B | 9 | −92.6 | 4.27E-4 | 7.0 | 0.1 |
Gm19219 | predicted gene, 19219 | 9 | −378.7 | 6.02E-3 | 3.8 | 0.0 |
Eif2s3y | eukaryotic translation initiation factor 2, subunit 3, Y-linked | Y | −3251.2 | 1.75E-37 | 5.6 | 0.0 |
Kdm5d | lysine (K)-specific demethylase 5D | Y | −4566.7 | 7.48E-42 | 2.2 | 0.0 |
Ddx3y | DEAD (Asp-Glu-Ala-Asp) box polypeptide 3, Y-linked | Y | −7263.9 | 7.91E-63 | 10.1 | 0.0 |
Analysis of pathways differentially impacted between male and female aged LECs at 24 hours PCS revealed that “response to Staphylococcus aureus infection”, metabolic pathways, and complement pathways were the most impacted (all at FDR≤ 0.04), while the DEGs mapped to the GO terms “cell adhesion” (FDR≤ 6 X10−5) and “extracellular matrix structural constituent” (FDR≤ 3 X10−9).
Comparison of genes differentially expressed between male and female aged LECs at 24 hours PCS, with those differentially expressed between young versus old LECs at 24 hours PCS when sex was not considered as a variable (Faranda et al 2021), revealed that five of the 430 genes differentially expressed by sex in aged LECs at 24 hours PCS were also expressed at significantly different levels in aged LECs at 24 hours PCS compared to young. In all cases, these five genes (Pik3ap1, Acod1, Ptafr, Hcar2, and Rpe65) are upregulated in both males and females, but the levels of all of these genes, except for Rpe65, are higher in aged male LECs than female at 24 hours PCS.
The effect of sex on the aged lens fiber cell transcriptome
Comparison between the lens fiber transcriptome of aged male and female mice revealed 41 biologically significant DEGs between the sexes, with 28 genes expressed at higher levels in females and 13 expressed at higher levels in males. Of these, five DEGs reside on sex chromosomes and 36 reside on autosomes (Table 7). Five DEGs located on sex chromosomes and 15 autosomal DEGs were previously found to exhibit sex dependent expression in other mouse tissues based on comparisons with SAGD (Table 7). Eight of the 41 biologically significant DEGs between male and female aged lens fibers exhibit lens preferred expression based on comparisons with the iSyTE database (Table 7).
Table 7.
DEGs between aged male and female lens fiber cells. Bold font denotes genes that have been previously reported to exhibit sex dependent expression in adult mouse tissues based on comparisons with SAGD (Shi et al., 2019). Italics font denotes genes that exhibit lens preferred expression in postnatal day 56 mouse lenses according to the iSyTe database (Kakrana et al., 2018).
SYMBOL | DESCRIPTION | Chromosome | Fold Difference | FDR | Male FPKM | Female FPKM |
---|---|---|---|---|---|---|
Xist | inactive X specific transcripts | X | 2931.0 | 8.13E-82 | 0.0 | 23.5 |
Prelp | proline arginine-rich end leucine-rich repeat | 1 | 25.0 | 4.15E-17 | 0.1 | 2.4 |
Crygd | crystallin, gamma D | 1 | 23.1 | 4.48E-2 | 40.2 | 929.7 |
Rbp3 | retinol binding protein 3, interstitial | 14 | 11.6 | 7.30E-3 | 0.4 | 4.4 |
H2bc3 | H2B clustered histone 3 | 13 | 7.4 | 4.58E-2 | 0.4 | 3.4 |
Ccnd2 | cyclin D2 | 6 | 5.9 | 4.21E-3 | 0.8 | 5.0 |
Gm4865 | predicted gene 4865 | 5 | 5.6 | 4.60E-2 | 0.6 | 3.2 |
Cxcl14 | chemokine (C-X-C motif) ligand 14 | 13 | 5.2 | 3.23E-5 | 0.9 | 5.1 |
Ndufa4l2 | Ndufa4, mitochondrial complex associated like 2 | 10 | 3.6 | 2.04E-2 | 2.9 | 10.6 |
Inhba | inhibin beta-A | 13 | 3.5 | 3.20E-2 | 1.7 | 5.8 |
S1pr3 | sphingosine-1-phosphate receptor 3 | 13 | 3.1 | 1.44E-8 | 1.6 | 5.2 |
Chrdl1 | chordin-like 1 | X | 3.1 | 5.19E-8 | 1.4 | 4.3 |
Pdpn | podoplanin | 4 | 2.9 | 1.40E-9 | 7.8 | 22.4 |
Aldh3a1 | aldehyde dehydrogenase family 3, subfamily A1 | 11 | 2.7 | 1.71E-5 | 53.1 | 143.0 |
Gpsm2 | G-protein signalling modulator 2 (AGS3-like, C. elegans) | 3 | 2.6 | 4.42E-2 | 2.6 | 6.9 |
Serping1 | serine (or cysteine) peptidase inhibitor, clade G, member 1 | 2 | 2.5 | 2.80E-3 | 1.6 | 4.1 |
Slc7a8 | solute carrier family 7 (cationic amino acid transporter, y+ system), 8 | 14 | 2.4 | 2.41E-4 | 5.9 | 14.4 |
Scarf2 | scavenger receptor class F, member 2 | 16 | 2.4 | 2.35E-2 | 2.0 | 4.8 |
Notch2 | notch 2 | 3 | 2.3 | 1.46E-2 | 2.2 | 5.1 |
Olfml2a | olfactomedin-like 2A | 2 | 2.3 | 1.63E-3 | 1.6 | 3.6 |
Kcnk1 | potassium channel, subfamily K, member 1 | 8 | 2.3 | 1.06E-2 | 3.0 | 6.8 |
Fez1 | fasciculation and elongation protein zeta 1 (zygin I) | 9 | 2.2 | 5.47E-3 | 1.7 | 3.8 |
F2r | coagulation factor II (thrombin) receptor | 13 | 2.2 | 3.71E-2 | 1.6 | 3.6 |
Msx1 | msh homeobox 1 | 5 | 2.2 | 4.92E-2 | 2.4 | 5.3 |
Susd2 | sushi domain containing 2 | 10 | 2.1 | 7.67E-3 | 1.9 | 4.1 |
Glul | glutamate-ammonia ligase (glutamine synthetase) | 1 | 2.1 | 6.38E-4 | 4.6 | 9.8 |
Mme | membrane metallo endopeptidase | 3 | 2.1 | 3.08E-3 | 2.8 | 5.7 |
Htra3 | HtrA serine peptidase 3 | 5 | 2.0 | 1.57E-2 | 4.7 | 9.5 |
Bpgm | 2,3-bisphosphoglycerate mutase | 6 | −2.1 | 7.15E-3 | 5.6 | 2.7 |
Guk1 | guanylate kinase 1 | 11 | −2.3 | 4.93E-2 | 8.6 | 3.7 |
Zfyve21 | zinc finger, FYVE domain containing 21 | 12 | −2.8 | 1.30E-2 | 7.7 | 2.8 |
Dmac1 | distal membrane arm assembly complex 1 | 4 | −3.4 | 3.23E-2 | 7.6 | 2.1 |
Igfbp2 | insulin-like growth factor binding protein 2 | 1 | −11.4 | 6.82E-4 | 4.2 | 0.4 |
Hba-a2 | hemoglobin alpha, adult chain 2 | 11 | −15.5 | 2.33E-4 | 63.2 | 4.1 |
Col8a2 | collagen, type VIII, alpha 2 | 4 | −23.7 | 7.32E-12 | 2.6 | 0.1 |
Hbb-bs | hemoglobin, beta adult s chain | 7 | −31.9 | 2.25E-3 | 28.7 | 0.9 |
Eif2s3y | eukaryotic translation initiation factor 2, subunit 3, Y-linked | Y | −31.9 | 6.15E-10 | 4.2 | 0.2 |
Alas2 | aminolevulinic acid synthase 2, erythroid | X | −43.2 | 2.55E-5 | 5.5 | 0.1 |
Krt12 | keratin 12 | 11 | −44.1 | 1.33E-3 | 3.6 | 0.1 |
Car3 | carbonic anhydrase 3 | 3 | −173.1 | 1.60E-3 | 6.4 | 0.0 |
Ddx3y | DEAD (Asp-Glu-Ala-Asp) box polypeptide 3, Y-linked | Y | −408.7 | 1.56E-60 | 7.5 | 0.0 |
Analysis of all DEGs that exhibited FDR corrected statistically significant levels of expression (233 genes total) for impacted pathways or enriched GO terms using iPathway guide (Ahsan and Draghici, 2017) revealed that “eye development” (19 genes total; FDR corrected p value ≤ 0.003) and extracellular matrix structural constituent (12 genes total; FDR ≤2.1 X10−4) were among the most enriched GO terms while the pathway “protein digestion and absorption was considered impacted at FDR ≤ 0.002.
Comparison between genes differentially expressed at biologically significant levels during aging in male lens fiber cells with those differentially expressed in aging female lens fibers revealed that 87 genes were upregulated in both sexes, while 324 were downregulated in both sexes, with no genes changing in opposite directions as a function of sex (See Supplemental Table 1). However, in many cases, the extent of age-related changes in gene expression was dimorphic between the sexes. Most notably, the age related decreases in γ-crystallin and other lens fiber cell marker mRNAs in lens fibers were generally more robust in males than females, while the age related upregulation of Ndufa4l2, a gene that encodes a protein that slows electron transport in damaged mitochondria (Li et al., 2017), is more robust in female lens fibers than male (Table 7; Supplemental Table 1).
Discussion
Aging is a biological process during which organisms undergo numerous physiological changes due to cellular senescence, along with age-related cell and tissue damage due to oxidative stress and other environmental insults (da Costa et al., 2016; Slawinska and Krupa, 2021; Xu et al., 2018). Individuals, and even tissues, age at different rates, which appears to be a function of both differential exposure to environmental insults and differing genetic and/or epigenetic landscape (Kenyon, 2010; Wood et al., 2015). The most robust known genetic influence on aging is sex chromosome complement, leading to sex-specific differences between the incidence/clinical presentation of numerous age-related diseases (Marquez et al., 2020; Mauvais-Jarvis et al., 2020; Sampathkumar et al., 2020).
In a companion study, we investigated the effect of age on the lens epithelial and fiber transcriptomes, as well as the response of LECs to cataract surgery, using C57BL/6J mice as the near genetic identity of inbred mice and uniform housing conditions reduces variability (Faranda et al., 2021). The young (12 week) old mice studied are sexually mature adults, while the aged (24 month old) mice appear biologically similar to 70 year old humans based on numerous physiological parameters (Marquez et al., 2020), including the observation that females have entered reproductive senescence based on parameters consistent with human menopause including elevations in serum follicle stimulating hormone (Collins et al., 1981; Urzua et al., 2017). As the prior study included equal numbers of male and female animals, here we were able to determine how sex influences the aging lens transcriptome.
Young LECs exhibit few sex differences in their transcriptomes although young lens fibers exhibit greater sexual dimorphism
Twelve week old mice are sexually mature, have transparent lenses, and have established their adult crystallin complement (Ueda et al., 2002). Transcriptome profiling of LECs from 12 week old mice isolated immediately after lens fiber cell removal revealed only five genes that exhibit at least a two-fold difference between sexes and were expressed at levels plausibly high enough to affect lens biology based on published criteria (Manthey et al., 2014a). One of these is Xist, a non-coding RNA critical for the inactivation of the second X chromosome in females (Brockdorff et al., 2020), while Ddx3y and Eif2s3y are neighboring genes found in the male specific region of the Y chromosome that have the potential to have diverse post transcriptional effects on gene expression (Deschepper, 2020). Neither autosomal DEG has a known function although one is predicted to be a non-coding RNA while the other may be protein coding. Similarly, young male and female LECs also remodel their transcriptome almost identically after a lens injury resembling cataract surgery, although three intriguing sex differences were also seen in aged LECs after injury. Ccdc88b encodes Gipie, a binding partner of GRP78 that can protect cells from the unfolded protein response (Matsushita et al., 2011) and G0s2, a regulator of lipid homeostasis (Zhang et al., 2017), are expressed at higher levels in young male LECs after injury. In contrast, Serping1, which encodes a serine protease inhibitor that regulates innate immunity (Davis et al., 2010), is expressed at higher levels in injured female LECs than male.
Young lens fibers have more transcriptomic differences between sexes than LECs, however, the detected fold changes tend to be modest, and bioinformatics analysis did not reveal significant potential biological connections between these DEGs. Interestingly though, two genes, Aldh3a1, which encodes an aldehyde dehydrogenase that detoxifies lipid peroxides and is known to protect the lens from stress induced opacity (Lassen et al., 2007), as well as Gstm1, which encodes glutathione S transferase mu, a xenobiotic detoxification enzyme which may be protective against cataract in humans (Saadat and Farvardin-Jahromi, 2006), are both expressed at higher levels in young female, compared to male lens fibers.
The lens cell transcriptome of aged mice exhibits greatly increased sexual dimorphism compared to young adults
We reported in the companion paper (Faranda et al., 2021) that both LECs and fibers downregulate the expression of numerous lens preferred genes, most notably the gamma crystallins, with aging, consistent with a prior study (Treton et al., 1988). Here we find that several of the crystallins that downregulate in lens cells with age appear to downregulate less dramatically in female lens cells than male, which correlates with male LECs expressing lower levels of lactase-like/klotho-gamma, a potential activator of FGF receptor activity in the lens (Audette et al., 2016). In contrast, aged male LECs express higher levels of Gpx3 mRNA than females. Notably, Gpx3 encodes a secreted glutathione peroxidase that detoxifies hydrogen peroxide in biological fluids (Baez-Duarte et al., 2014; Olson et al., 2010), suggesting that aged male lenses could be better protected against the oxidative damage than females. This proposal is supported by the observation that aged female lens fibers express higher levels of Ndufa4l2, a marker of mitochondrial damage (Li et al., 2017), than males. These intriguing results could suggest a mechanism for the epidemiological observation that post menopausal women are more prone to developing cataract than men of similar age (Zetterberg and Celojevic, 2015).
The most dramatic sexual dimorphism in lens gene expression detected in this study was for LECs isolated from aged eyes 24 hours after they underwent a surgery that models cataract removal (Manthey et al., 2014b). Pathway analysis of these DEGs revealed that genes encoding several ECM molecules as well components of the complement cascade were differentially expressed in female LECs at 24 hours PCS compared to males. Interestingly, a recent study suggested that Xist, which is a highly female preferred transcript in all lens cells, can increase ECM mRNA levels in injured skin cells by sponging microRNAs that mediate ECM mRNA degradation (Cao and Feng, 2019), an effect that may enhance fibrotic disease progression (Wang et al., 2017). Further, sex differences in complement pathway regulation have been previously reported in C57BL/6 mice (Kotimaa et al., 2016) and normal humans (Gaya da Costa et al., 2018; Troldborg et al., 2017), and such dimorphisms are implicated in differences in disease prevalence, severity, and outcome between sexes (Mauvais-Jarvis et al., 2020; Ward et al., 2018). While it is currently unknown how early transcriptome remodeling of LECs after lens fiber cell removal (Jiang et al., 2018) mechanistically relates to the pathogenesis of posterior capsular opacification (PCO), it is tempting to speculate that sex differences in complement activation and ECM production explain the increased PCO prevalence in women observed in some epidemiological studies (Congdon et al., 2008; Fong et al., 2014; Lee et al., 2016).
Limitations of this study
While the present study and the companion paper (Faranda et al, 2021) revealed that the lens transcriptome differs as a function of sex and age, the limitations of the study design also indicate the need to explore how sex affects lens biology in more depth using both unbiased approaches and directed investigation. First, it is possible, if not likely, that sex has a greater effect on the lens transcriptome than suggested in this study. The limited availability of 24 month old inbred mice housed under controlled conditions led to the present study design where only two biological replicates could be analyzed for sex (male versus female) under each condition (young versus old epithelium, fibers, and injured epithelium). The resulting low statistical power to detect differences in gene expression could mean that there are additional sex-dependent effects on the lens transcriptome, although this issue is likely minimized due to the study of inbred mice housed in a controlled environment. Second, while RNAseq is very reproducible, we were unable to obtain aged animals for validation studies due to their limited availability. Third, it will be important to replicate this work in humans to ensure that these differences have biological relevance to human cataract. Fourth, the broader effects of sex on PCO development are still open questions. The epidemiological evidence for differential PCO rates between sexes is currently mixed (Congdon et al., 2008; Fong et al., 2014; Lee et al., 2016; Tokko et al., 2019) , while many studies on PCO incidence either do not consider sex dependence (Haripriya et al., 2017) or the study design controls for sex (Chen et al., 2019), so sex differences are obscured. Fifth, the present study investigated the acute inflammatory response of mouse LECs to fiber cell removal, at a time 1-2 days prior to the phenotypic transition of LECs to myofibroblasts (Shihan et al., 2020), so the transcriptomic differences by sex detected here may not reflect sex differences in the LEC fibrotic response. Sixth, the animal model used here does not strictly reproduce the surgical technique (including intraocular lens placement) used in human cataract surgery. Thus, future studies are needed to investigate both whether sex influences how robust the later fibrotic response of LECs to lens injury is, the long term survival of these myofibroblasts, and their migratory potential. The latter two points are critical to translate this work to humans as clinical PCO usually occurs months or years after cataract surgery when lens cells initially trapped at the capsular bag periphery by IOL interactions with the lens capsule (Bisevac et al., 2020) escape their containment, and migrate into the optical axis.
Implications for lens biology
The risk of developing age-related cataract in humans is influenced by sex and age (Chang et al., 2011; Hugosson and Ekstrom, 2020), with epidemiological studies routinely finding that post-menopausal females have elevated risk of cataract development compared to aged matched males. While it has been hypothesized that estrogen withdrawal at menopause has direct negative effects on the lens (Zetterberg and Celojevic, 2015), the present study did not find appreciable expression of canonical estrogen receptor mRNA in adult mouse lens cells. However, aged lens cells did exhibit sexually dimorphic expression of numerous genes, several of which have the potential to modify cataract risk. It will be interesting to determine whether such age-related sex differences in gene expression also exist in humans, the mechanisms by which this sexual dimorphism is established, their functional relevance to lens biology, and ultimate risk of cataract development.
Implications for experimental design in lens and cataract research
Similar to the lens, others have found that tissue transcriptomes commonly differ between sexes, with numerous autosomal genes exhibiting sex-preferred expression. However, autosomal genes often only exhibit sexually dimorphic expression in a few, or even a single tissue, demonstrating that chromosomal sex has tissue specific effects (Arnold, 2019; Kassam et al., 2019; Lu and Mar, 2020; Mukaibo et al., 2019; Oliva et al., 2020; Shi et al., 2019). This observation also is valid for the lens, as few autosomal genes exhibiting sexually dimorphic expression in the lens exhibit sexually dimorphic expression in other mouse tissues based on comparisons with the SAGD database (Shi et al., 2019).
These observations, along with increasing recognition that sex confers differences in disease risk/pathogenesis and drug metabolism, has led the National Institutes of Health (NIH) of the United States to mandate that sex be considered as a biological variable in NIH supported biomedical research (Woitowich et al., 2020). Overall, the present study supports the need to both include animals of both sexes in lens research, and to be aware of the potential for sex to modify lens biology, particularly during investigations of lens aging and its influence on cataract.
Supplementary Material
Highlights:
Young lens epithelial cells exhibit few sex dependent differences in gene expression
Young lens epithelial cells exhibit few sex dependent differences in their response to lens injury
Young lens fibers exhibit numerous sex dependent differences in gene expression
Aged lens epithelial cells exhibit modest numbers of sexually dimorphic genes
Aged lens epithelial cells exhibit profound sex specific responses to lens injury
Aged lens fiber cells exhibit increased numbers of genes that exhibit sexually dimorphic gene expression
Funding:
This study was funded by grants from the National Eye Institute (EY028597 and EY028597-S1). The 24 month old C57BL/6/Nia mice used in this study were a gift from the United States National Institute on Aging, Division of Aging Biology, Biological Resources Branch under exception approval Duncan091818. Support from the University of Delaware CBCB Bioinformatics Core Facility and use of the BIOMIX compute cluster was made possible through funding from Delaware INBRE (NIH NIGMS P20 GM103446), the State of Delaware, and the Delaware Biotechnology Institute.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Ackert-Bicknell CL, Anderson LC, Sheehan S, Hill WG, Chang B, Churchill GA, Chesler EJ, Korstanje R, Peters LL, 2015. Aging Research Using Mouse Models. Curr Protoc Mouse Biol 5, 95–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahsan S, Draghici S, 2017. Identifying Significantly Impacted Pathways and Putative Mechanisms with iPathwayGuide. Curr Protoc Bioinformatics 57, 7 15 11–17 15 30. [DOI] [PubMed] [Google Scholar]
- Anders S, Pyl PT, Huber W, 2014. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arnold AP, 2017. A general theory of sexual differentiation. J Neurosci Res 95, 291–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arnold AP, 2019. Rethinking sex determination of non-gonadal tissues. Current topics in developmental biology 134, 289–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Audette DS, Anand D, So T, Rubenstein TB, Lachke SA, Lovicu FJ, Duncan MK, 2016. Prox1 and fibroblast growth factor receptors form a novel regulatory loop controlling lens fiber differentiation and gene expression. Development 143, 318–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baez-Duarte BG, Mendoza-Carrera F, Garcia-Zapien A, Flores-Martinez SE, Sanchez-Corona J, Zamora-Ginez I, Torres-Rasgado E, Leon-Chavez BA, Perez-Fuentes R, Multidisciplinary Research Group on Diabetes of the Instituto Mexicano del Seguro, S., 2014. Glutathione peroxidase 3 serum levels and GPX3 gene polymorphisms in subjects with metabolic syndrome. Arch Med Res 45, 375–382. [DOI] [PubMed] [Google Scholar]
- Bisevac J, Anisimova NS, Nagymihaly R, Kristianslund O, Katta K, Noer A, Sharafetdinov IH, Drolsum L, Moe MC, Malyugin BE, Petrovski G, 2020. Long-term myofibroblast persistence in the capsular bag contributes to the late spontaneous in-the-bag intraocular lens dislocation. Sci Rep 10, 20532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brockdorff N, Bowness JS, Wei G, 2020. Progress toward understanding chromosome silencing by Xist RNA. Genes Dev 34, 733–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Call MK, Grogg MW, Del Rio-Tsonis K, Tsonis PA, 2004. Lens regeneration in mice: implications in cataracts. Exp Eye Res 78, 297–299. [DOI] [PubMed] [Google Scholar]
- Cao W, Feng Y, 2019. LncRNA XIST promotes extracellular matrix synthesis, proliferation and migration by targeting miR-29b-3p/COL1A1 in human skin fibroblasts after thermal injury. Biol Res 52, 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang JR, Koo E, Agron E, Hallak J, Clemons T, Azar D, Sperduto RD, Ferris FL 3rd, Chew EY, Age-Related Eye Disease Study, G., 2011. Risk factors associated with incident cataracts and cataract surgery in the Age-related Eye Disease Study (AREDS): AREDS report number 32. Ophthalmology 118, 2113–2119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen HC, Lee CY, Sun CC, Huang JY, Lin HY, Yang SF, 2019. Risk factors for the occurrence of visual-threatening posterior capsule opacification. J Transl Med 17, 209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y, Lun A, Smyth G, 2016. From reads to genes to pathways: differential expression analysis of RNA-Seq experiments using Rsubread and the edgeR quasi-likelihood pipeline [version 2; peer review: 5 approved]. F1000Research 5. [DOI] [PMC free article] [PubMed]
- Collins TJ, Parkening TA, Smith ER, 1981. Plasma and pituitary concentrations of LH, FSH and prolactin after injection of GnRH in aged female C57BL/6 mice. Neurobiol Aging 2, 125–131. [DOI] [PubMed] [Google Scholar]
- Congdon N, Fan H, Choi K, Huang W, Zhang L, Zhang S, Liu K, Hu IC, Zheng Z, Lam DS, 2008. Impact of posterior subcapsular opacification on vision and visual function among subjects undergoing cataract surgery in rural China: Study of Cataract Outcomes and Up-Take of Services (SCOUTS) in the Caring is Hip Project, report 5. Br J Ophthalmol 92, 598–603. [DOI] [PubMed] [Google Scholar]
- da Costa JP, Vitorino R, Silva GM, Vogel C, Duarte AC, Rocha-Santos T, 2016. A synopsis on aging-Theories, mechanisms and future prospects. Ageing Res Rev 29, 90–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis AE 3rd, Lu F, Mejia P, 2010. C1 inhibitor, a multi-functional serine protease inhibitor. Thromb Haemost 104, 886–893. [DOI] [PubMed] [Google Scholar]
- Desai VD, Wang Y, Simirskii VN, Duncan MK, 2010. CD44 expression is developmentally regulated in the mouse lens and increases in the lens epithelium after injury. Differentiation 79, 111–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deschepper CF, 2020. Regulatory effects of the Uty/Ddx3y locus on neighboring chromosome Y genes and autosomal mRNA transcripts in adult mouse non-reproductive cells. Sci Rep 10, 14900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Draghici S, Khatri P, Tarca AL, Amin K, Done A, Voichita C, Georgescu C, Romero R, 2007. A systems biology approach for pathway level analysis. Genome Res 17, 1537–1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Faranda Adam P., Shihan Mahbubul H., Wang Yan, and Duncan Melinda K. (2021) The aging mouse lens transcriptome Experimental eye research 209:108663. doi: 10.1016/j.exer.2021.108663 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fong CS, Mitchell P, Rochtchina E, Cugati S, Hong T, Wang JJ, 2014. Three-year incidence and factors associated with posterior capsule opacification after cataract surgery: The Australian Prospective Cataract Surgery and Age-related Macular Degeneration Study. Am J Ophthalmol 157, 171–179 e171. [DOI] [PubMed] [Google Scholar]
- Gaya da Costa M, Poppelaars F, van Kooten C, Mollnes TE, Tedesco F, Wurzner R, Trouw LA, Truedsson L, Daha MR, Roos A, Seelen MA, 2018. Age and Sex-Associated Changes of Complement Activity and Complement Levels in a Healthy Caucasian Population. Front Immunol 9, 2664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorin MB, Day R, Costantino JP, Fisher B, Redmond CK, Wickerham L, Gomolin JE, Margolese RG, Mathen MK, Bowman DM, Kaufman DI, Dimitrov NV, Singerman LJ, Bornstein R, Wolmark N, 1998. Long-term tamoxifen citrate use and potential ocular toxicity. Am J Ophthalmol 125, 493–501. [DOI] [PubMed] [Google Scholar]
- Greenfield A, 2015. Understanding sex determination in the mouse: genetics, epigenetics and the story of mutual antagonisms. J Genet 94, 585–590. [DOI] [PubMed] [Google Scholar]
- Haripriya A, Chang DF, Vijayakumar B, Niraj A, Shekhar M, Tanpreet S, Aravind S, 2017. Long-term Posterior Capsule Opacification Reduction with Square-Edge Polymethylmethacrylate Intraocular Lens: Randomized Controlled Study. Ophthalmology 124, 295–302. [DOI] [PubMed] [Google Scholar]
- Hodes GE, Walker DM, Labonte B, Nestler EJ, Russo SJ, 2017. Understanding the epigenetic basis of sex differences in depression. J Neurosci Res 95, 692–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hugosson M, Ekstrom C, 2020. Prevalence and risk factors for age-related cataract in Sweden. Ups J Med Sci 125, 311–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang J, Shihan MH, Wang Y, Duncan MK, 2018. Lens Epithelial Cells Initiate an Inflammatory Response Following Cataract Surgery. Invest Ophthalmol Vis Sci 59, 4986–4997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kakrana A, Yang A, Anand D, Djordjevic D, Ramachandruni D, Singh A, Huang H, Ho JWK, Lachke SA, 2018. iSyTE 2.0: a database for expression-based gene discovery in the eye. Nucleic Acids Res 46, D875–D885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K, 2017. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res 45, D353–D361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kassam I, Wu Y, Yang J, Visscher PM, McRae AF, 2019. Tissue-specific sex differences in human gene expression. Hum Mol Genet 28, 2976–2986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kenyon CJ, 2010. The genetics of ageing. Nature 464, 504–512. [DOI] [PubMed] [Google Scholar]
- Kim D, Paggi JM, Park C, Bennett C, Salzberg SL, 2019. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol 37, 907–915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kotimaa J, Klar-Mohammad N, Gueler F, Schilders G, Jansen A, Rutjes H, Daha MR, van Kooten C, 2016. Sex matters: Systemic complement activity of female C57BL/6J and BALB/cJ mice is limited by serum terminal pathway components. Mol Immunol 76, 13–21. [DOI] [PubMed] [Google Scholar]
- Lassen N, Bateman JB, Estey T, Kuszak JR, Nees DW, Piatigorsky J, Duester G, Day BJ, Huang J, Hines LM, Vasiliou V, 2007. Multiple and additive functions of ALDH3A1 and ALDH1A1: cataract phenotype and ocular oxidative damage in Aldh3a1(−/−)/Aldh1a1(−/−) knock-out mice. J Biol Chem 282, 25668–25676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JS, Chung CC, Lin KK, Yu KH, Kuo CF, See LC, 2016. Time trends in cataract surgery and after-cataract laser capsulotomy in Taiwan: A population-based retrospective cohort study. Int J Surg 36, 265–273. [DOI] [PubMed] [Google Scholar]
- Li J, Bai C, Guo J, Liang W, Long J, 2017. NDUFA4L2 protects against ischaemia/reperfusion-induced cardiomyocyte apoptosis and mitochondrial dysfunction by inhibiting complex I. Clin Exp Pharmacol Physiol 44, 779–786. [DOI] [PubMed] [Google Scholar]
- Lowe R, Gemma C, Rakyan VK, Holland ML, 2015. Sexually dimorphic gene expression emerges with embryonic genome activation and is dynamic throughout development. BMC Genomics 16, 295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu T, Mar JC, 2020. Investigating transcriptome-wide sex dimorphism by multi-level analysis of singlecell RNA sequencing data in ten mouse cell types. Biol Sex Differ 11, 61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manthey AL, Terrell AM, Lachke SA, Polson SW, Duncan MK, 2014a. Development of novel filtering criteria to analyze RNA-sequencing data obtained from the murine ocular lens during embryogenesis. Genom Data 2, 369–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manthey AL, Terrell AM, Wang Y, Taube JR, Yallowitz AR, Duncan MK, 2014b. The Zeb proteins δEF1 and Sip1 may have distinct functions in lens cells following cataract surgery. Invest Ophthalmol Vis Sci 55, 5445–5455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marquez EJ, Chung CH, Marches R, Rossi RJ, Nehar-Belaid D, Eroglu A, Mellert DJ, Kuchel GA, Banchereau J, Ucar D, 2020. Sexual-dimorphism in human immune system aging. Nat Commun 11, 751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matsushita E, Asai N, Enomoto A, Kawamoto Y, Kato T, Mii S, Maeda K, Shibata R, Hattori S, Hagikura M, Takahashi K, Sokabe M, Murakumo Y, Murohara T, Takahashi M, 2011. Protective role of Gipie, a Girdin family protein, in endoplasmic reticulum stress responses in endothelial cells. Mol Biol Cell 22, 736–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mauvais-Jarvis F, Bairey Merz N, Barnes PJ, Brinton RD, Carrero JJ, DeMeo DL, De Vries GJ, Epperson CN, Govindan R, Klein SL, Lonardo A, Maki PM, McCullough LD, Regitz-Zagrosek V, Regensteiner JG, Rubin JB, Sandberg K, Suzuki A, 2020. Sex and gender: modifiers of health, disease, and medicine. Lancet 396, 565–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mukaibo T, Gao X, Yang NY, Oei MS, Nakamoto T, Melvin JE, 2019. Sexual dimorphisms in the transcriptomes of murine salivary glands. FEBS Open Bio 9, 947–958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nelson HD, Fu R, Zakher B, Pappas M, McDonagh M, 2019. Medication Use for the Risk Reduction of Primary Breast Cancer in Women: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA 322, 868–886. [DOI] [PubMed] [Google Scholar]
- Noran NH, Salleh N, Zahari M, 2007. Relationship between reproductive exposures and age-related cataract in women. Asia Pac J Public Health 19, 23–28. [DOI] [PubMed] [Google Scholar]
- Oliva M, Munoz-Aguirre M, Kim-Hellmuth S, Wucher V, Gewirtz ADH, Cotter DJ, Parsana P, Kasela S, Balliu B, Vinuela A, Castel SE, Mohammadi P, Aguet F, Zou Y, Khramtsova EA, Skol AD, Garrido-Martin D, Reverter F, Brown A, Evans P, Gamazon ER, Payne A, Bonazzola R, Barbeira AN, Hamel AR, Martinez-Perez A, Soria JM, Consortium G.T., Pierce BL, Stephens M, Eskin E, Dermitzakis ET, Segre AV, Im HK, Engelhardt BE, Ardlie KG, Montgomery SB, Battle AJ, Lappalainen T, Guigo R, Stranger BE, 2020. The impact of sex on gene expression across human tissues. Science 369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olson GE, Whitin JC, Hill KE, Winfrey VP, Motley AK, Austin LM, Deal J, Cohen HJ, Burk RF, 2010. Extracellular glutathione peroxidase (Gpx3) binds specifically to basement membranes of mouse renal cortex tubule cells. Am J Physiol Renal Physiol 298, F1244–1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phipson B, Lee S, Majewski IJ, Alexander WS, Smyth GK, 2016. Robust hyperparameter estimation protects against hypervariable genes and improves power to detect differential expression. The Annals of Applied Statistics 10, 946–963, 918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robinson MD, McCarthy DJ, Smyth GK, 2010. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robinson MD, Oshlack A, 2010. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol 11, R25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saadat M, Farvardin-Jahromi M, 2006. Occupational sunlight exposure, polymorphism of glutathione S-transferase M1, and senile cataract risk. Occup Environ Med 63, 503–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sampathkumar NK, Bravo JI, Chen Y, Danthi PS, Donahue EK, Lai RW, Lu R, Randall LT, Vinson N, Benayoun BA, 2020. Widespread sex dimorphism in aging and age-related diseases. Hum Genet 139, 333–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi MW, Zhang NA, Shi CP, Liu CJ, Luo ZH, Wang DY, Guo AY, Chen ZX, 2019. SAGD: a comprehensive sex-associated gene database from transcriptomes. Nucleic Acids Res 47, D835–D840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shihan MH, Kanwar M, Wang Y, Jackson EE, Faranda AP, Duncan MK, 2020. Fibronectin has multifunctional roles in posterior capsular opacification (PCO). Matrix Biol. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Slawinska N, Krupa R, 2021. Molecular Aspects of Senescence and Organismal Ageing-DNA Damage Response, Telomeres, Inflammation and Chromatin. Int J Mol Sci 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith RS, Roderick TH, Sundberg JP, 1994. Microphthalmia and associated abnormalities in inbred black mice. Lab Anim Sci 44, 551–560. [PubMed] [Google Scholar]
- Storey JD, Tibshirani R, 2003. Statistical significance for genomewide studies. Proc Natl Acad Sci U S A 100, 9440–9445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tarca AL, Draghici S, Khatri P, Hassan SS, Mittal P, Kim JS, Kim CJ, Kusanovic JP, Romero R, 2009. A novel signaling pathway impact analysis. Bioinformatics 25, 75–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tokko HA, Hussain F, Al-Awadi A, Mei F, Zeiter JH, Kim C, Tannir JR, Shukairy A, Juzych MS, McDermott ML, Nassiri N, Hughes BA, 2019. Factors Associated with the Development of Posterior Capsule Opacification Requiring Yttrium Aluminum Garnet Capsulotomy. Optom Vis Sci 96, 492–499. [DOI] [PubMed] [Google Scholar]
- Treton JA, Jacquemin E, Courtois Y, 1988. Variation in the relative abundance of gamma-crystallin gene transcripts during development and ageing. Exp Eye Res 46, 405–413. [DOI] [PubMed] [Google Scholar]
- Troldborg A, Hansen A, Hansen SW, Jensenius JC, Stengaard-Pedersen K, Thiel S, 2017. Lectin complement pathway proteins in healthy individuals. Clin Exp Immunol 188, 138–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ueda Y, Duncan MK, David LL, 2002. Lens proteomics: the accumulation of crystallin modifications in the mouse lens with age. Invest Ophthalmol Vis Sci 43, 205–215. [PubMed] [Google Scholar]
- Urzua U, Chacon C, Lizama L, Sarmiento S, Villalobos P, Kroxato B, Marcelain K, Gonzalez MJ, 2017. Parity History Determines a Systemic Inflammatory Response to Spread of Ovarian Cancer in Naturally Aged Mice. Aging Dis 8, 546–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y, Liang Y, Luo J, Nie J, Yin H, Chen Q, Dong J, Zhu J, Xia J, Shuai W, 2017. XIST/miR-139 axis regulates bleomycin (BLM)-induced extracellular matrix (ECM) and pulmonary fibrosis through beta-catenin. Oncotarget 8, 65359–65369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ward LJ, Olausson P, Li W, Yuan XM, 2018. Proteomics and multivariate modelling reveal sex-specific alterations in distinct regions of human carotid atheroma. Biol Sex Differ 9, 54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woitowich NC, Beery A, Woodruff T, 2020. A 10-year follow-up study of sex inclusion in the biological sciences. Elife 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wood SH, van Dam S, Craig T, Tacutu R, O’Toole A, Merry BJ, de Magalhaes JP, 2015. Transcriptome analysis in calorie-restricted rats implicates epigenetic and post-translational mechanisms in neuroprotection and aging. Genome Biol 16, 285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, Inman CL, Ogrodnik MB, Hachfeld CM, Fraser DG, Onken JL, Johnson KO, Verzosa GC, Langhi LGP, Weigl M, Giorgadze N, LeBrasseur NK, Miller JD, Jurk D, Singh RJ, Allison DB, Ejima K, Hubbard GB, Ikeno Y, Cubro H, Garovic VD, Hou X, Weroha SJ, Robbins PD, Niedernhofer LJ, Khosla S, Tchkonia T, Kirkland JL, 2018. Senolytics improve physical function and increase lifespan in old age. Nat Med 24, 1246–1256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yates AD, Achuthan P, Akanni W, Allen J, Allen J, Alvarez-Jarreta J, Amode MR, Armean IM, Azov AG, Bennett R, Bhai J, Billis K, Boddu S, Marugan JC, Cummins C, Davidson C, Dodiya K, Fatima R, Gall A, Giron CG, Gil L, Grego T, Haggerty L, Haskell E, Hourlier T, Izuogu OG, Janacek SH, Juettemann T, Kay M, Lavidas I, Le T, Lemos D, Martinez JG, Maurel T, McDowall M, McMahon A, Mohanan S, Moore B, Nuhn M, Oheh DN, Parker A, Parton A, Patricio M, Sakthivel MP, Abdul Salam AI, Schmitt BM, Schuilenburg H, Sheppard D, Sycheva M, Szuba M, Taylor K, Thormann A, Threadgold G, Vullo A, Walts B, Winterbottom A, Zadissa A, Chakiachvili M, Flint B, Frankish A, Hunt SE, G II, Kostadima M, Langridge N, Loveland JE, Martin FJ, Morales J, Mudge JM, Muffato M, Perry E, Ruffier M, Trevanion SJ, Cunningham F, Howe KL, Zerbino DR, Flicek P, 2020. Ensembl 2020. Nucleic Acids Res 48, D682–D688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Younan C, Mitchell P, Cumming RG, Panchapakesan J, Rochtchina E, Hales AM, 2002. Hormone replacement therapy, reproductive factors, and the incidence of cataract and cataract surgery: the Blue Mountains Eye Study. Am J Epidemiol 155, 997–1006. [DOI] [PubMed] [Google Scholar]
- Zetterberg M, Celojevic D, 2015. Gender and cataract--the role of estrogen. Curr Eye Res 40, 176–190. [DOI] [PubMed] [Google Scholar]
- Zhang X, Heckmann BL, Campbell LE, Liu J, 2017. G0S2: A small giant controller of lipolysis and adipose-liver fatty acid flux. Biochim Biophys Acta Mol Cell Biol Lipids 1862, 1146–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao F, Yao HH, 2019. A tale of two tracts: history, current advances, and future directions of research on sexual differentiation of reproductive tractsdagger. Biol Reprod 101, 602–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.