Research Paper
Exploring the Stability of Genomic Imprinting and X-Chromosome Inactivation in the Aged Brain
Authors
Samantha Mancino,1,2,3,¶ Janith Seneviratne,4,5,¶ Annalisa Mupo,6,7 Felix Krueger,7,8 David Oxley,9 Melanie A. Eckersley-Maslin,4,5,10,* and Simão Teixeira da Rocha1,2,3,*
1Instituto de Bioengenharia e Biociências e Departamento de Bioengenharia, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
2Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
3Faculdade de Medicina, Instituto de Medicina Molecular, João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
4Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
5Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
6Epigenetics Programme, Babraham Institute, Cambridge, United Kingdom
7Altos Labs, Cambridge, United Kingdom
8Bioinformatics Group, Babraham Institute, Cambridge, United Kingdom
9Mass Spectrometry Facility, The Babraham Institute, Cambridge, United Kingdom
10Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, Australia
*Corresponding authors: melanie.eckersley-maslin@petermac.org; simao.rocha@tecnico.ulisboa.pt
¶Samantha Mancino and Janith Seneviratne have equally contributed to this research.
DOI:https://doi.org/10.59368/agingbio.20240030
Received: 9/29/2023, Revised: 4/11/2024, Accepted: 6/22/2024, Published: 7/31/2024
Full Article | PDF | Supplementary
Abstract
Changes in the epigenetic landscape are a hallmark of aging that contributes to the irreversible decline in organismal fitness ultimately leading to aging-related diseases. Epigenetic modifications regulate the cellular memory of the epigenetic processes of genomic imprinting and X-chromosome inactivation (XCI) to ensure monoallelic expression of imprinted and X-linked genes. Whether aging-associated epigenetic changes affect the maintenance of genomic imprinting and XCI has not been comprehensively studied. Here, we investigate the allele-specific transcriptional and epigenetic signatures of the aging brain, by comparing juvenile and old hybrid mice obtained from C57BL/6J (BL6) and CAST/EiJ (CAST) reciprocal crosses, with an emphasis on the hippocampus (HCP). We confirmed that the aged HCP showed an expected increase in DNA hydroxymethylation and a typical aging transcriptional signature. Importantly, genomic imprinting was largely unaffected, with stable parent-of-origin-specific DNA methylation in multiple brain regions including the HCP, cerebellum, nucleus accumbens, hypothalamus, and prefrontal cortex. Consistently, allele-specific transcriptomic bulk analysis confirmed unaltered imprinting expression in the aged HCP. An exception was four novel non-coding transcripts (B230209E15Rik, Ube2nl, A330076H08Rik, and A230057D06Rik) at the Prader-Willi syndrome/Angelman syndrome imprinted locus, which lost strict monoallelic expression during aging. Similar to imprinting, XCI was remarkably stable with no signs of aging-driven skewing or relaxation of monoallelic expression of X-linked genes. Our study provides a valuable resource for evaluating monoallelic expression in the aging brain and reveals that, despite the known epigenetic changes occurring during aging, genomic imprinting and XCI remain predominantly stable throughout the process of physiological aging in the mouse brain.
Introduction
Aging can be defined as an irreversible loss of physiological integrity associated with the functional decline of tissues and organs, progressively leading to aging-related illnesses, such as neurodegenerative diseases1. At the molecular and cellular levels, several hallmarks have been associated with aging, including changes in the epigenetic landscape2,3. DNA methylation is affected by these aging-related epigenetic changes. Here, cytosine Bases followed by guanine, known as CpG sites, can acquire a methyl group at the C-5 position (5-methylcytosine [5mC]). Some regions of the genome can undergo age-induced gains and losses in DNA methylation, and these changes can condition their pattern of gene expression4. This pattern of DNA methylation changes during lifespan can be used as an “epigenetic clock” to predict chronological age5,6. This phenomenon indicates that challenges in preserving epigenetic marks may lead to changes in accessibility and gene expression patterns that, in turn, impact the cellular and molecular functions of aged cells5.
Neurons, as long-lived post-mitotic cells in the brain, are characterized by an evolving epigenetic landscape during differentiation, maturation, and aging, making them a prime target for studying cellular aging in the context of brain function and health7–11. Disease signatures based on DNA methylation patterns have also been linked to a variety of age-related neurological and psychiatric disorders12–15. 5mC can be catalyzed to 5-hydroxymethylcytosine (5hmC) by the ten-eleven translocation (Tet) family of dioxygenases as a part of the active DNA demethylation cycle. While much less abundant than 5mC, 5hmC levels are particularly high in the adult brain when compared with other somatic tissues10 and accumulate during aging16–18. The precise significance of 5hmC in the brain and its accumulation during lifespan has been postulated to result from enhanced DNA demethylation activity necessary for the epigenetic regulation of brain-specific genes involved in neurodevelopmental processes and neuronal function and plasticity19. The dynamic changes in 5mC and 5hmC in the brain are illustrative of the different layers of complex epigenetic regulation used by neurons and other brain cells to integrate signals and outputs underlying highly skillful processes such as learning and memory.
Epigenetic mechanisms, such as DNA methylation, are main actors in the regulation of monoallelic expression of genes, playing a crucial role in the establishment and/or maintenance of the mammalian epigenetic processes of genomic imprinting and X-chromosome inactivation (XCI)20. Many genes regulated by these processes have critical roles in brain development and function and are thought to contribute to the diversity and specialization of neuronal cells21. Whether aging-associated epigenetic changes affect the heritability of monoallelic expression during physiological aging and impact the aging process remains an open question.
Imprinted genes consist of a unique subset of ∼150 genes displaying parental-of-origin-specific gene expression. The majority are located in ∼25 genomic clusters where their monoallelic expression is dependent on DNA methylation at CpG-dense regulatory regions, known as imprinting control regions (ICRs)22. This DNA methylation is asymmetrically deposited during female and male germline development. Interestingly, a substantial number of imprinted genes exhibit monoallelic or biased expression from one parental allele in one tissue or at specific developmental stage23,24. In this regard, the brain is one of the organs where more genes show tissue-, isoform-, or developmental-stage-specific imprinting25–27. Within the brain, this is highly regionalized, with different areas exhibiting their own set of monoallelic or parentally biased expressed genes26,28. The importance of imprinted genes in brain function is evidenced by the devastating neurological and behavioral conditions such as Angelman and Prader-Willi syndromes resulting from (epi)mutations affecting the chr15q11-q13 region in humans29. Transcriptomic studies have shown that imprinting expression in the cerebellum (CB) is developmentally regulated25. Whether imprinting is also susceptible to changes as a function of aging has not been systematically addressed.
XCI is a dosage compensation mechanism that equalizes X-linked gene expression of XX females to XY males30. This process is established early in development and is regulated by the X-inactive-specific transcript (XIST) long non-coding RNA (lncRNA). During embryogenesis, XIST is upregulated randomly from one of the two X chromosomes and becomes exclusively expressed from the inactive X chromosome (Xi). This lncRNA engages in a complex interplay with several RNA-binding proteins to recruit transcriptional repressors and chromatin modifiers, establishing the silenced state of the Xi31,32. Although the molecular mechanisms underlying the initiation of XCI have been extensively elucidated, our understanding of the long-term maintenance of XCI throughout an organism’s lifespan remains limited. Recent investigations in aging, with a focus on the hematopoietic cell lineage, reveal an escalation in XCI skewing, where one parental allele is preferentially inactivated33 along with subtle alterations in DNA methylation patterns and gene expression across the X chromosome34,35. Interestingly, a separate study conducted in the brain, employing single nuclei transcriptomics, unveiled an intriguing finding: Xist expression is observed to be upregulated in aged neurons located within the hypothalamus and hippocampus (HCP) of female mice36. The implications of this observation on XCI remain ambiguous, underscoring the necessity for in-depth investigations to elucidate the influence of aging on XCI within the context of the brain.
In the present study, we provide the first allele-specific epigenetic and transcriptional landscape of the aging mouse brain. We particularly focus on the HCP, a key regulator brain area of cognitive processes that tend to decline during aging. We used juvenile (8–9 weeks) and old (>100 weeks) F1 hybrid mice from reciprocal crosses between distantly related mouse strains to gain allelic resolution. DNA methylation and transcriptomic analysis enabled us to discern the impact of aging on monoallelic expression in the brain. Our results support the stable epigenetic inheritance of genomic imprinting and XCI during physiological aging of the mouse brain.
Materials and Methods
Ethics
Animal welfare and experimental procedures were conducted according to the ethical guidelines of the European Directive 2010/63/EU and the Portuguese legislation DL 113/2013 and were approved by the responsible Ethical Committee of Instituto de Medicina Molecular João Lobo Antunes (iMM) and the Portuguese competent authority, Direção Geral de Alimentação e Veterinária (license number 023357/19).
Animals
Mice colonies of Mus musculus C56BL/6J (BL6) strain and Mus musculus castaneus CAST/EiJ (CAST) strain were obtained from the Jackson Laboratory and maintained at the iMM Rodent facility. Animals were housed in a maximum of five per cage in a temperature- and humidity-controlled room (24°C, 45%–65%) with a 14/12 hour light/dark cycle. Animals were fed diet ad libitum.
Reciprocal crosses between BL6 and CAST animals—BL6/CAST (BL6 female and CAST male) and CAST/BL6 (CAST female and BL6 male) were established to generate F1 animals. Juvenile and old F1 animals were developed and sacrificed by cervical dislocation in the range of 8–9 weeks and 102–104 weeks, respectively. A total of 10 young female (six BL6/CAST and four CAST/BL6), four young male (two BL6/CAST and two CAST/BL6), seven old female (five BL6/CAST and two CAST/BL6), and five old male (two BL6/CAST and three CAST/BL6) were used in this study (Table S1). Female mice were not synchronized for the estrous cycle.
Samples preparation
Sacrificed animals were decapitated by cervical dislocation. The brains were quickly removed and the whole CB and hypothalamus were rapidly isolated from the brainstem. The following brain areas were then dissected according to the atlas of stereotaxic coordinates of mouse brain37 and immersed in liquid nitrogen for 4 sec: HCP (from bregma, Anterior/Posterior [AP]: from −1.34 to −2.56 mm; Medial/Lateral [ML]: ±0 mm; Dorsal/Ventral [DV]: −3 mm), medial prefrontal cortex (from bregma, AP: from −2.10 to −1.70 mm; ML: ±0 mm; DV: −3.5 mm), and nucleus accumbens (from bregma, AP: from −1.54 to −0.98 mm; ML: ±0.5 mm; DV: −4 mm). Lung tissue was also collected. After collection, brain areas and lung tissue were immediately frozen in liquid nitrogen and stored at −80°C for later molecular analysis. DNA and RNA were isolated for each brain area or lung tissues from the selected animal tissues using the NucleoSpinTriPrep kit (Cat# 740966.50, Macherey-Nagel GmbH & Co.KG, Germany) according to the manufacturer’s guidelines.
5mC/5hmC measurements by liquid chromatography–mass spectrometry (LC-MS/MS)
Genomic DNA from the CB, HCP, and lung of both juvenile and old female mice was digested using DNA Degradase Plus (Cat# E2020, Zymo Research) according to the manufacturer’s instructions. Nucleosides were analyzed by LC-MS/MS on a Q-Exactive mass spectrometer (Thermo Scientific) fitted with a nanoelectrospray ion source (Proxeon). All samples and standards had a heavy isotope-labeled nucleoside mix added prior to mass spectral analysis (2′-deoxycytidine-13C1, 15N2 [Cat# SC-214045, Santa Cruz], 5-(methyl-2H3)-2′-deoxycytidine [Cat# SC-217100, Santa Cruz], 5-(hydroxymethyl)-2′-deoxycytidine-2H3 [Cat# H946632, Toronto Research Chemicals]). MS2 data for 5hmC, 5mC, and C were acquired with both the endogenous and corresponding heavy-labeled nucleoside parent ions simultaneously selected for fragmentation using a 5 Th isolation window with a 1.5 Th offset. Parent ions were fragmented by higher-energy collisional dissociation with a relative collision energy of 10% and a resolution setting of 70,000 for MS2 spectra. Peak areas from extracted ion chromatograms of the relevant fragment ions, relative to their corresponding heavy isotope-labeled internal standards, were quantified against a six-point serial twofold dilution calibration curve, with triplicate runs for all samples and standards.
Bisulfite treatment
Genomic DNA (1 μg) from the CB, HCP, hypothalamus, medial prefrontal cortex, nucleus accumbens, and lung of four juvenile and four old female mice (two animals for each reciprocal cross) was bisulfite converted using the EZ DNA methylation Gold kit (Cat# D5006, Zymo Research) according to the manufacturer’s instructions. After column cleanup, the DNA was eluted in an elution buffer (66 μl) to obtain a final concentration of ∼15 ng/μl bisulfite converted DNA.
IMPLICON library preparation and analysis
IMPLICON was performed as previously described38 for the CB, HCP, nucleus accumbens, prefrontal cortex, hypothalamus, and lung of four juvenile and four old female mice (two animals for each reciprocal cross). Briefly, following bisulfite conversion, a first polymerase chain reaction (PCR) amplifies each region per sample in individual reactions, adding adapter sequences, as well as eight random nucleotides (N8) for subsequent data deduplication. PCR conditions and primers for this first step are listed in Table S2. Primers cover 11 imprinted clusters (10 ICRs and exon1a promoter of Ddc gene), together with two unmethylated (Sox2, Klf4) and one methylated (Prickle1) control regions. After pooling amplicons for each biological sample and clean-up using AMPure XP magnetic beads (Cat# A63880, Beckman Coulter), a second PCR completes a sequence-ready library with sample barcodes for multiplexing. In this PCR reaction, barcoded Illumina adapters are attached to the pooled PCR samples ensuring that each sample pool receives a unique reverse barcoded adapter. Libraries were verified by running 1:30 dilutions on an Agilent bioanalyzer and then sequenced using the Illumina MiSeq platform to generate paired-end 250 bp reads using the indexing primer with the following sequence, 5′-AAGAGCGGTTCAGCAGGAATGCCGAGACCGATCTC-3′ and 10% PhIX spike-in because the libraries are low complexity. We run two independent IMPLICON libraries that were named: first run (lane 7651) and second run (lane 7950). The first run contained the CB, HCP, nucleus accumbens, prefrontal cortex, hypothalamus, and lung of four juvenile and four old female mice, while the second run contained the samples of CB, HCP, and lung of the same mice.
IMPLICON bioinformatics analysis was also performed as described previously38, following the step-by-step guide of data processing analysis in https://github.com/FelixKrueger/IMPLICON. Briefly, data were processed using standard Illumina base-calling pipelines. As the first step of processing, the first 8 bp of Read 2 were removed and written into the readID of both reads as an in-line barcode or unique molecular identifier (UMI). This UMI was then later used during the deduplication step with “deduplicate bismark barcode mapped_file.bam.” Raw sequence reads were then trimmed to remove both poor-quality calls and adapters using Trim Galore v0.5.0 (www.bioinformatics.babraham.ac.uk/projects/trim_galore/, Cutadapt version 1.15, parameters: --paired). Trimmed reads were aligned with the mouse reference genome in paired-end mode. Alignments were carried out with Bismark v0.20.0. CpG methylation calls were extracted from the mapping output using the Bismark methylation extractor. Deduplication was then carried out with deduplicate_bismark using the barcode option to take UMIs into account (see above). The data were aligned with a hybrid genome of BL6/CAST (the genome was prepared with the SNPsplit package v0.3.4 [https://github.com/FelixKrueger/SNPsplit]). Following alignment and deduplication, reads were split allele specifically with SNPsplit. Aligned read (.bam) files were imported into Seqmonk software v1.47 (http://www.bioinformatics.babraham.ac.uk/projects/seqmonk) for all downstream analysis. Probes were made for each CpG contained within the amplicon and quantified using the DNA methylation pipeline or total read count options. Downstream analysis was performed using Microsoft Excel spreadsheet software (v2206 Build 16. 0. 15330. 20144) and GraphPad Prism v8.0.1.
From the raw data deposited in gene expression omnibus (GEO) under the accession number GSE148067, the reads mapped to the following murine (mm10) genomic coordinates were excluded from consideration in this article for one of the following reasons: (1) regions that fail to reach the coverage threshold for the two parental alleles in a given sample (>50 reads), including 3 of 13 imprinted regions, Igf2-H19, Igf2r, and Grb10, presented in our original IMPLICON primer set38; (2) regions sequenced twice for which only the run with more reads was considered; and (3) regions out of the scope of this article. For the samples sequenced in lane 7651, this includes: Chr7:60005043-60005284, Chr7:142581761-142582087, Chr12:109528253-109528471, Chr6:30737609-30737809, Chr11:12025411-12025700, and Chr18:12972868-12973155; for the samples sequenced in lane 7950, this includes: Chr7:142581761-142582087, Chr7:142659774-142664092, and Chr11:12025411-12025700.
RNA sequencing
The quality of DNAse I-treated total RNA from female young and old hippocampi (n = 5 old mice: n = 2 CAST-BL6 and n = 3 BL6-CAST; n = 6 young mice, n = 3 of each reciprocal cross) was checked by 2100 Agilent Bioanalyser. Samples with RNA integrity number (RIN) score above 9 were processed. RNA (1 μg) was used as input for PolyA+ directional RNA-seq library preparation using the NEBNext Ultra II Directional RNA-seq Kit (#E7765, NEB) with the PolyA mRNA magnetic isolation module (#E7490, NEB) according to the manufacturer’s instructions. The pooled library was sequenced on an Illumina HiSeq 2000 with a 2 x 100 bp kit.
Fastqs were processed using TrimGalore v0.6.6 in paired-end mode with default parameters. Validated read pairs were then aligned with the GRCm38.v5 mouse genome using Hisat2 v2.1.0 with the following parameters: --dta, --no-softclip, --no-mixed, and --no-discordant. The resulting hits were filtered to remove mappings with Mapping Quality (MAPQ) scores of < 20 and then converted to BAM format using Samtools v1.10. Allele-specific alignments were also performed by realigning mapped reads to N-masked genomes C56BL/6 J (genome 1) and CAST/EiJ (genome 2), which was based on the GRCm38.v5 genome and generated using the SNPsplit v0.3.4 package. Reads that were then sorted by allele-specificity for either genome or reads containing conflicting Single Nucleotide Polymorphism (SNP) information were excluded. Total and allele-specific read counts were quantified from BAMs with feature counts (from the subread v2.0.0 package) using default parameters with gencode vM25 annotations. Basic statistics on read counts and mappability are provided in Table S3.
We used SNP information to measure allele-specific expression (ASE) and performed DESeq2 analysis between maternal and paternal alleles, excluding those genes that exhibited random (i.e., genes with monoallelic expression independent of parental origin or strain) or strain-dependent (i.e., all genes with biased expression according to the genetic background) monoallelic expression.
DESeq2 v1.34.0 package in an R (v4.1.2)/R Studio (v2022.02.0+443) environment was used thus for conducting all differential gene expression analyses (including total and allele-specific analyses). For all analyses (including gene level), low expressed genes were first filtered (genes with ≥ 10 read counts across both alleles in each sample and ≥ 1 TPM in each sample across all samples were kept). For allele-specific analyses, allelic ratios were first derived by dividing the maternal or paternal allele counts by the total number of allelic counts (ratios are between 0 and 1). These allelic ratios were then used as input into DESeq2 after adjusting size factors to 1 for each sample (to account for allelic ratio input). To determine ASE (either maternal or paternal) across all mice, a DESeq design (∼0 + age + genome + sample + allele) was used with blocking terms against age group and the genome of origin for each allele to reduce the impact of age- or cross-specific allelic expression. To determine ASE (either maternal or paternal) in young and old mice separately, a separate DESeq2 design (∼0 + genome + age + age:sample + age:allele) was used. Contrasts were then made between old and young mice to determine age-specific and ASE changes that were attributed to age. Genes with ASE were considered as those with an absolute log2FoldChange > 1 and adjusted p-value (Benjamini–Hochberg adjusted) < 0.05. For calculating the proportion of reads aligning to the mouse genome, aligned reads on ChrX were counted from BAMs using feature counts either in an allele-specific context (post-SNPsplit for BL6 and CAST alleles) or allele-independent context (pre-SNPsplit). These counts were then divided by the total number of aligned reads and multiplied by 106 to obtain ChrX reads per million. ChrX count proportions for the BL6 allele were determined by dividing BL6 counts on ChrX by the total read counts on ChrX across both alleles (post-SNPsplit).
Principal component analysis (PCA) was conducted using the prcomp function from the stats v4.1.2 R package using DESeq2 normalized counts for all genes. Barplots, boxplots, and volcano plots were plotted using the ggplot2 v3.3.5 R package, and heatmaps were constructed using the ComplexHeatmap v2.10.0 R package. The genomic distribution of imprinted genes on chromosomes was plotted using the karyoplotR v1.20.3 R package. Track plots of normalized read densities (for RNA-seq data) were plotted for several genomic loci of interest using the rtracklayer v1.54.0 and Gviz v1.38.4 R packages.
For the analysis of the bulk HCP dataset from Hahn et al.39, raw gene counts were obtained from GEO under the accession number GSE212336. This dataset was subsetted for samples originating from female mice hippocampi (both posterior and anterior sites) for comparison with the data generated in this study. In this comparison, young mice were considered to be 3 months old, while old mice were considered to be 21 months old. These are the timepoints that better match our young (∼2 months) and old (∼24 months) animals. We performed differential gene expression analyses between old and young mice in this dataset using DESeq2 with a design (∼anatomical_site + age) and the same thresholds for statistical significance as above was considered (absolute log2FoldChange > 1, BH-adjusted p-value < 0.05). Heatmaps for these data were plotted for various genes using ComplexHeatmap, wherein DESeq2 normalized counts were subject to z-score normalization across samples within the same tissue type prior to plotting (note for the heatmap, animals of 12, 15, and 18 months old from Hahn’s dataset were also considered). To summarize gene expression signatures (upregulated and downregulated), the mean of z-score normalized values across the relevant genes for each signature was taken. Column graphs for various genes were plotted for these data using ggplot2.
CIBERSORTx analysis
Single-cell RNA-seq datasets in the form of UMI count matrices were retrieved for the hippocampi of four mice (two young and two old) from GEO under the accession number GSE16134040. Matrices were loaded and processed using the Seurat v4.2.041 R package (Read10X > CreateSeuratObject > merge). The fraction of reads aligned to mitochondrial (mt-) or ribosomal (Rps|Rpl|Mrps|Mrpl) genes out of all reads was calculated for each cell using PercentageFeatureSet. Cells were quality filtered for the UMI counts (100 < n < 20000), number of genes expressed (200 < n < 6000), mitochondrial read (<0.1), and ribosomal read (<0.5) fractions. Data were then subjected to normalization and scaling using SCTransform with the top 3000 variable genes. PCAs (RunPCA), mutual nearest neighbors (FindNeighbors, top 30 principal components), Uniform Manifold Approximation and Projection (UMAP) dimensional reduction (RunUMAP), and clustering (FindClusters, resolution = 1) were then performed. Gene signatures were constructed using canonical cell type marker genes defined in a comparative scRNA-seq study of mouse hippocampi in the literature42. Module scores that summarize the gene expression of canonical gene signatures among cell types were then computed using AddModuleScore. Violin plots were plotted using VlnPlot. Cell type classifications were made per cell cluster, wherein median cell type signature scores surpassing 0 were used to assign a given cell type. For deconvolution analyses, each cell type was randomly downsampled to 100 cells (sample and subset). Downsampled raw UMI count matrices with cell type annotations were further subsetted for genes commonly expressed in our bulk RNA-seq dataset. This matrix was then input to CIBERSORTx43 (https://cibersortx.stanford.edu/) as a single-cell reference matrix, wherein a signature matrix was first derived to determine those genes that accurately predicted each cell type (replicates [100], sampling [0.5], and fraction [0.0]). Cell fractions were then imputed for the HCP samples derived in this study (raw counts) using the aforementioned signature matrix with CIBERSORTx, without batch correction. Imputed cell fractions for each sample were then plotted using the ggplot2 R package. Cell type proportions in old and young mice were compared using pairwise t-tests (BH adjusted to account for multiple comparisons).
We also analyzed single-cell RNA-seq data for Fluorescence-Activated Cell Sorting (FACS)-sorted whole mouse brains (both myeloid and non-myeloid cell types) from the Tabula Muris Senis study, which profiled several whole young and old mouse brains44. The pre-processed RNA-seq counts containing 17 cell type annotations for cell types identified in brain tissues contained within the TabulaMurisSenisData R package (v1.0.0) were used as input to CIBERSORTx to first derive a signature matrix (replicates [15], sampling [0.5], and fraction [0.0]). Cell fractions were then imputed for the HCP samples derived in this study with the aforementioned signature matrix using CIBERSORTx.
Statistics
Statistical analysis used for each experiment is indicated in the respective figure legend with p-values indicated or marked as *p-value < 0.05, **p < 0.01, and ***p < 0.001. According to the distribution of data analyzed by the Shapiro–Wilk test, the following tests for the differential analysis of the experiments were used: unpaired two-tailed Welch’s t-test (
Results
Increase in 5hmC levels is a hallmark of the aging HCP
To decipher the allele-specific epigenetic and transcriptional features of the aging brain, we established reciprocal crosses between BL6 and CAST mice to generate female and male BL6-CAST and CAST-BL6 F1 hybrid mice (
We first evaluated the overall levels of 5mC and 5hmC by LC-MS/MS in young and old tissues from female and male animals of both reciprocal crosses (Table S1). 5mC levels did not differ significantly among CB, HPC, and lung and were not affected by age (
Transcriptomic signatures of the aged HCP
We next examined alterations in the transcriptome occurring during aging of the HCP by employing RNAseq on samples obtained from both young and old mice. We selected the HCP due to the increase in 5hmC levels associated with aging. Specifically, we performed RNAseq analysis of female HCP, comprising a total of five samples from aged mice (two CAST-BL6 and three BL6-CAST individuals) and six samples from young mice (three individuals from each reciprocal cross). Clustering using PCA did not distinguish between mice of opposite reciprocal crosses, yet the combination of PC1 and PC2 was able to separate old and young mice (Fig. S2A). This dataset was thoroughly scrutinized to assess shifts in the expression of genes associated with the DNA methylation machinery over the aging process as well as to investigate the broader patterns of gene expression changes.
To better understand the cause leading to the increase in 5hmC levels upon aging, we first analyzed expression levels of the Tet methylcytosine dioxygenases (Tet1, Tet2, and Tet3) responsible for the sequential conversion of 5mC to 5hmC, and subsequent oxidation steps as a part of the active DNA demethylation pathway47. No differences were observed for any of the three Tet genes by RNAseq in HCP upon aging (
Next, to conduct a broader analysis of gene expression, we performed differential gene expression analysis that identified 75 upregulated and 115 downregulated genes during aging process (adjusted p-value [P adj] < 0.05, absolute log2 fold change [FC] > 1) (
We validated the transcriptomic differences between young and old HCP by comparing it with a comprehensive dataset that profiled 847 brain samples (spanning 15 anatomical regions and taken from mice of varying ages between 3 and 28 months)39. Of relevance to our study were samples from the anterior and posterior HCP from 3, 12, 15, 18, and 21 months old female mice. We compared the expression of all DEGs between young and old HCP in our study in the anterior and posterior HCP from these female mice and observed gene expression trends that aligned with the direction of our DEGs, as shown by average z-score plots (Fig. S2B). We next specifically assessed DEGs between 3- and 21-month-old samples in this dataset to assess the level of overlap with our dataset. For this analysis, anterior and posterior HCP samples were analyzed together to increase the power of comparisons between age groups and not to miss any pan-HCP changes that we likely observe in our dataset. There were 44 up- and 2 downregulated genes (adjusted p-value < 0.05 and absolute log2FC > 1) in the Hahn et al. dataset39 of which 13 upregulated genes (Lyz2, C4b, Spag6, Lcn2, Bcl3, Cd22, Itgax, Onecut1, Ccl3, AA414992, Upk1b, H2-Q7, and Pcdhb2) and 1 downregulated gene (Eomes) were similarly differentially expressed in our dataset (Table S5), indicating that 30.4% of the DEGs in the Hahn dataset39 were also identified in our analysis. This overlap was statistically significant (Fisher’s exact test using all commonly expressed genes [n = 20960] between our study and Hahn et al.’s study as the background [up: p-value = 2.3e−22, down: p-value = 0.011]). Of note, the hippocampal samples in Hahn et al.39 are divided into the anterior/posterior regions, whereas the hippocampal samples in our study are from the whole HCP. These differences in anatomical sites could account for unique gene expression in each dataset.
To address the potential impact differences in cell type proportions between individual mice in our bulk transcriptomes, we estimated cell proportions using a cell type deconvolution algorithm. CIBERSORTx models (see the
To complement this analysis, we performed GSEA to investigate further the cellular processes altered with age (Table S6). GSEA identified four major cellular processes that were altered upon age. Downregulated genes were enriched for cell cycle, cell signaling, and neurodevelopment processes, while upregulated genes were enriched for immune response and inflammation (
Imprinting methylation is stable during aging
Next, we investigated how aging influences the fidelity of genomic imprinting. This was made possible due to the use of reciprocal BL6xCAST crosses to discern parent-of-origin from genetic effects25,45. We first screened for imprinting methylation using IMPLICON: an amplicon sequencing method measuring DNA methylation at several ICRs across the genome at the nucleotide resolution with high coverage38. With this method, we are also able to separate out paternal from maternal reads based on SNPs between BL6 and CAST strains that are contained in our amplicons and conserved during bisulfite conversion. IMPLICON was successfully performed on 11 imprinted clusters (10 ICRs and the exon1a promoter of Ddc gene) together with two unmethylated and one methylated control regions (
Unmethylated (Klf4 and Sox2) and methylated (Prickle1) controls showed low (<∼10%) or high (>∼90%) DNA methylation levels, respectively, at both maternal and paternal alleles for all tissues analyzed (Fig. S3A; Table S7), irrespective of age. Importantly, DNA methylation at all ICRs analyzed was stably maintained with age in the HCP, CB, and lung (Table S7). This is illustrated for the maternally methylated Peg3 and Plagl1 loci (
We also examined DNA methylation consistency between individual CpGs along individual reads using the IMPLICON method. Reads were either fully unmethylated or methylated depending on their parent of origin, independent of the genetic background. This is exemplified for Peg3 and Plagl1 ICRs in HCP (
Using IMPLICON, we also checked the methylation levels of the Dopa decarboxylase (Ddc) gene, which is involved in dopamine biosynthesis, and often dysregulated in neurodegenerative and psychiatric disorders49. Ddc transcripts are generally expressed from both parental alleles in most of the body tissues and can also exhibit isoform-, cell-, and development-specific imprinted expression, including in the brain50,51. In our samples, the exon1a promoter of the imprinted gene isoform of Ddc was not differentially methylated between the parental alleles, nor throughout aging, in HCP and CB, with methylation levels in two brain areas being lower than in the lung (∼50% vs. 90%) (Fig. S3Aiii).
Finally, we also extended our IMPLICON analysis to three additional brain regions: the nucleus accumbens, prefrontal cortex, and hypothalamus. Similar to CB and HCP, these regions also showed high fidelity of differential DNA methylation for the six imprinting clusters and the control regions analyzed (Table S8). In conclusion, our results suggest that imprinting methylation is stably maintained during aging for the loci and brain areas investigated in this study.
The “Imprintome” of the young and old HCP
We next determined the “imprintome” of the aging mouse HPC by taking advantage of our transcriptome dataset from F1 mice of reciprocal crosses. In total, we identified 113 genes with parental-of-origin monoallelic expression in young HCP, of which 66 were maternally and 49 were paternally expressed. These genes were located in 17 genomic regions that corresponded to known imprinted regions (
Consistent with our IMPLICON results, RNAseq analysis revealed stable imprinting expression during aging in HCP. For example, Peg3, Usp29, Zim3, and Plagl1 genes are exclusively paternally expressed in young and old HCP (
More globally, when comparing old versus young ASE for imprinted genes using DESeq2 method, we found four not previously reported imprinted genes (B230209E15Rik, Ube2nl, A330076H08Rik, and A230057D06Rik) showing aging-specific partial erosion of strict pattern of monoallelic expression (deviating from a maternal:paternal ratio of ∼0: ∼100% in young to ∼25:∼75% in old HPC) (
XCI in the aging brain
XCI is a dosage compensation mechanism that silences one of the two X chromosomes in female mammalian cells31. We used our RNAseq dataset, which includes young and old female F1 hybrid mice from reciprocal crosses, to examine the status of XCI during aging process in HCP. First, we assessed the expression levels of the long noncoding RNA Xist that is the master regulator of XCI. There were no differences in Xist levels between young and old HCP (
Discussion
In this study, we present the first allele-specific epigenetic and transcriptional landscape of the aging mouse brain, with a specific emphasis on the HCP. Using a combination of IMPLICON and RNA sequencing analyses, our findings highlight the preservation of genomic imprinting and the stability of XCI throughout the natural aging process in the murine brain.
An epigenetic signature of aging has been implicated in the irreversible decline of organismal fitness and the onset of aging-related illnesses57. Notably, epigenetic modifications, including DNA methylation, undergo predictable changes over time, enabling the estimation of biological age based on DNA methylation “epigenetic clocks”58. Whether these epigenetic-induced changes affect maintenance of monoallelic expression including genomic imprinting and XCI was not known and thus was the focus of this study.
One typical epigenetic feature associated with the aging brain is the increase in 5hmC levels over time59. Indeed, 5hmC levels increase markedly during lifespan, suggesting that 5hmC-mediated epigenetic modification may be critical in neurodevelopment and neurodegenerative disorders60,61. Our data confirmed that 5hmC levels are higher in the brain than in peripheral tissues and increases in the HCP with age, irrespective of biological sex60,62 (
Our transcriptome analysis identified both up- and downregulated transcripts in the aged HCP. Notably, we found higher expression of genes related to inflammation and immune responses, while genes involved in cellular cycle progression, neurodevelopmental processes, and active signaling pathways displayed reduced expression levels (
Our CIBERSORTx deconvolution analysis using two independent single-cell datasets40,44 (Fig. S2C) revealed that the neural population of our hippocampal samples was notably high (∼90%–95%) and did not vary in proportion between young and old HCP. There was a small increase in astrocyte proportions when using the single-cell dataset of the aging whole brain from The Tabula Muris Consortium as the reference44 (right panel, Fig. S2C). Future studies with larger datasets will enable gene expression estimations for major cell types estimated by CIBERSORTx, providing a deeper understanding of cell-type-specific gene expression changes. Together, our transcriptome analysis aligns with previous studies36,75 and matches other aging RNAseq datasets40,44, suggesting a consistent and generalized pattern of gene expression changes associated with aging.
Genomic imprinting is an enduring form of epigenetic inheritance established in parental germ cells and maintained throughout an organism’s development24. In the central nervous system, imprinting is important for neurogenesis, brain function, and behavior27 and dysregulation of imprinting results in neurodevelopmental and behavioral disorders such as PWS/AS24,29. In accordance, the brain, especially neurons, consistently shows a high number of expressed imprinted genes in adulthood18,76,77. A detailed investigation of imprinted gene expression in the aging HCP was yet to be performed, despite the previous association between imprinting methylation and hippocampal volume in aging78. Recent evidence suggested that imprints can be dysregulated by environmental insults during critical periods of embryonic/fetal development79–81 with long-term consequences in tissue function and susceptibility to age-related diseases25,76. Whether imprinting in the brain is also susceptible to changes as a function of aging has not been addressed previously in a systematic way.
Using our allele-specific IMPLICON method, we observed a consistent DNA methylation pattern across 11 imprinted regions in the HCP, CB, and lung tissues of both young and old F1 mice as well as in their reciprocal crosses. However, we also noted that certain CpGs exhibited either increased or decreased methylation. Whether this is attributed to experimental noise or indeed has biological meaning would require further investigation. Because IMPLICON used bisulfite treatment, it cannot distinguish between 5mC and 5hmC, and so we were unable to address the potential impact of the aging-specific increase in 5hmC on imprinted regions. To tease apart 5mC and 5hmC levels at imprinting or other regions, novel techniques can now be applied such as Tet-assisted bisulfite sequencing (TAB-seq)82 and oxidative bisulfite sequencing (oxBS-seq)83 or emerging direct detection and single-cell methods82 to start addressing the potential role of 5hmC at imprinted regions during aging.
We also assessed age-related allele-specific transcriptional changes in the HCP in our RNAseq dataset, documenting 113 coding and non-coding genes with parental allelic expression in the HCP across aging. In line with our IMPLICON findings, imprinted gene expression remained stable throughout the aging process. While we cannot rule out dysregulation of imprinting in the context of age-onset diseases or following environmental insults84,85, our findings show stable parental-of-origin DNA methylation and transcription at imprinted loci during physiological aging of the brain. One caveat in our analysis is the absence of single-cell resolution, which masks the existence of other imprinted genes that may exhibit cell-type-specific imprinting. This may indeed be the case of Ddc gene belonging to the Grb10 imprinted locus on chr11, which we postulated to undergo transcriptional or imprinting regulation upon aging and thus influencing dopamine production50,51,86. Single-cell techniques, such as scRNAseq, would be required to disclose the full “imprintome” of HCP during aging. Bulk RNA-seq, performed by us in this study, remains the gold standard in studying global trends in monoallelic expression in a wide range of different contexts25 due to its reproducibility, scalability, and cost effectiveness. However, it cannot uncover more subtle changes unique to specific cell types or states. Over the past few decades, the landscape of single-cell RNA-seq (scRNA-seq) methods has experienced rapid evolution with constant improvement in sensitivity, throughput, and reproducibility. For accurately quantifying ASE in single cells, well-based methods (e.g., Smart-seq2/3) have been preferred87. For instance, ASE quantification by scRNA-seq has been used to elucidate the degree of XCI escape across human tissues88, the dynamics of X-chromosome silencing89, or to unravel dosage compensation mechanisms in mammalian preimplantation development90. Importantly, scRNA-seq will be crucial to understand the true extent of ASE in different cell types and states87. Technological improvements, including long-read single-cell RNAseq, will help overcome current challenges in ASE quantification using scRNA-seq including limitations in analyzing low-abundance transcripts, sequencing drop outs, and scalability limitations. Alternatively, cell sorting of different cell populations followed by bulk RNA sequencing has the potential to reveal subtle changes in allelic expression during aging that may be specific to particular cell types. In particular, this would enable detection of allelic expression changes that occur in opposite directions in different cell types91, which might otherwise be obscured by using whole tissue preparations as we did in this study.
Although our data strongly point for an enduring stability of genomic imprinting in the aging brain, an exception was found for four novel non-coding transcripts at the PWS/AS imprinted locus on mouse chr7 that lost strict monoallelic expression upon aging: B230209E15Rik, Ube2nl, A330076H08Rik, and A230057D06Rik. These transcripts are strongly expressed in brain tissues; however, their exact function is currently unknown. The most enigmatic of them all is Ube2nl, an intronless pseudogene with an open reading frame derived from the Ube2n (ubiquitin-conjugating enzyme E2N) gene on mouse chr10. While we observed consistent allele biases changes in the aged HCP for these genes, this did not translate into overall gene expression changes. Functional studies will be needed to understand the role of these transcripts in mouse development and aging and whether the syntenic region in humans is also sensitive to aging-specific effect on imprinting regulation.
We also investigated changes in XCI during aging process and did not observe any differences in our dataset (
Our study represents a comprehensive investigation of the allele-specific DNA methylation and transcriptional landscape of the aging brain. While we focused on genomic imprinting and XCI, this dataset holds promise for exploring other monoallelic-specific phenomena that may be relevant to aging. We acknowledge the limitations in our study including the exclusive focus on female mice and the relatively small cohort of animals. This decision stemmed from ethical considerations and was justified as a reduced number of mice are needed when working with an isogenic setting where all animals share an identical genotype in controlled conditions. It is worth noting that although our F1 hybrid mice from reciprocal crosses could be seen as a subgroup within the young and old groups, our findings (Figs. S1B, S3B, and S4A,S4B) indicate that they did not differ within these age groups. Hence, we were able to have n = 4 young/old animals for the IMPLICON and n >= 5 young/old animals for the RNAseq analysis. Comparable numbers of mice have been used in similar aging studies (∼5 female mice in Hahn et al.
In conclusion, our findings support the remarkable resilience of genomic imprinting and XCI in the face of epigenetic changes observed during healthy aging of the brain, providing advances beyond what has been previously achieved. The stability of these epigenetic processes with aging suggests their importance in preserving essential cellular functions throughout an entire individual’s lifespan.
Acknowledgments
We would like to thank the members of the S.T.d.R.’s team for helpful discussions and the animal facility of the iMM for their help in maintaining the animal colonies. Work in S.T.d.R.’s team was supported by Fundação para a Ciência e Tecnologia (FCT) Ministério da Ciência, Tecnologia e Ensino Superior (MCTES), Portugal (IC&DT projects 2022.01532.PTDC and PTDC/BIA-MOL/29320/2017 as well as projects UIDB/04565/2020 and UIDP/04565/2020 of the Research Unit Institute from Bioengineering and Biosciences—iBB and LA/P/0140/2020 of the Associate Laboratory Institute for Health and Bioeconomy—i4HB) and S.T.d.R. and S.M. were supported by assistant research contracts from FCT/MCTES (2021.00660.CEECIND and CEECIND/02356.2021, respectively). Research in M.A.E.-M.’s laboratory was supported by a Snow Medical Fellowship awarded to M.A.E.-M. and the Lorenzo and Pamela Galli Medical Research Trust.
Author Contributions
S.T.d.R. conceived the study, supervised the project, and together with M.A.E.-M. secured funding. S.M. maintained the BL6 and CAST mouse lines, sacrificed the mice, dissected brain areas, performed most of the molecular biology experiments, conducted IMPLICON experiments, analyzed 5mC/5hmC measurements, IMPLICON, and RNA-seq results. J.S. prepared the RNAseq alignment, analyzed RNA-seq data, and the CYBERSORTx deconvolution analysis under the supervision of M.A.E.-M. D.O. performed the 5mC/5hmC measurements. F.K. and M.A.E.-M. conducted bioinformatic analysis of IMPLICON. A.M. helped in RNAseq, IMPLICON, and LC-MS/MS experiments. S.M. and S.T.d.R. wrote the article with contributions of J.S. and M.A.E.-M.
Conflicts of Interests
A.M. and F.K. are Altos Labs employees. The other authors declare no competing interests.
Data Availability
GEO superseries (RNAseq and IMPLICON) can be accessed here: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE232548.
Supplementary Materials
Supplemental information can be found here: Supplementary.