Targeting the hypothalamus for modeling age-related dna methylation and developing oxt-gnrh combinational therapy against alzheimer’s disease-like pathologies in male mouse model

Targeting the hypothalamus for modeling age-related dna methylation and developing oxt-gnrh combinational therapy against alzheimer’s disease-like pathologies in male mouse model


Play all audios:


ABSTRACT The hypothalamus plays an important role in aging, but it remains unclear regarding the underlying epigenetics and whether this hypothalamic basis can help address aging-related


diseases. Here, by comparing mouse hypothalamus with two other limbic system components, we show that the hypothalamus is characterized by distinctively high-level DNA methylation during


young age and by the distinct dynamics of DNA methylation and demethylation when approaching middle age. On the other hand, age-related DNA methylation in these limbic system components


commonly and sensitively applies to genes in hypothalamic regulatory pathways, notably oxytocin (OXT) and gonadotropin-releasing hormone (GnRH) pathways. Middle age is associated with


transcriptional declines of genes which encode OXT, GnRH and signaling components, which similarly occur in an Alzheimer’s disease (AD)-like model. Therapeutically, OXT-GnRH combination is


substantially more effective than individual peptides in treating AD-like disorders in male 5×FAD model. In conclusion, the hypothalamus is important for modeling age-related DNA methylation


and developing hypothalamic strategies to combat AD. SIMILAR CONTENT BEING VIEWED BY OTHERS NONCANONICAL REGULATION OF IMPRINTED GENE _IGF2_ BY AMYLOID-BETA 1–42 IN ALZHEIMER’S DISEASE


Article Open access 04 February 2023 FTO-MEDIATED M6A DEMETHYLATION REGULATES GNRH EXPRESSION IN THE HYPOTHALAMUS VIA THE PLCΒ3/CA2+/CAMK SIGNALLING PATHWAY Article Open access 21 December


2023 CORTICOSTERONE INDUCES DISCRETE EPIGENETIC SIGNATURES IN THE DORSAL AND VENTRAL HIPPOCAMPUS THAT DEPEND UPON SEX AND GENOTYPE: FOCUS ON METHYLATED _NR3C1_ GENE Article Open access 16


March 2022 INTRODUCTION Aging-related neurodegenerative diseases such as AD have become major health problems among the elderly, but clinical solutions are still missing partly due to the


incomplete knowledge of this disease. Interestingly, recent research has suggested that the functioning of neurogenic brain regions in early life could have long-term effects on


aging-related neuronal degeneration and loss1,2,3,4. As is known, there are three major types of neurogenic brain regions, including the hippocampal dentate gyrus5,6, the subventricular


zone7,8,9,10, and the mediobasal hypothalamus11,12,13, which are responsible for adult neurogenesis related to the limbic system functions of the hippocampus, olfactory bulb (OB), and


hypothalamus, respectively. Recently, our research has led to an understanding that the hypothalamus plays a regulatory role in systemic aging and systemic physiology11,12,13,14,15, while


the underlying basis involves the actions from hypothalamic neuropeptides such as GnRH and OXT15,16,17,18. However, to date there is still a lack of research comparing the hypothalamus and


other limbic system components in the context of addressing aging or aging-related diseases. To fill this gap, we designed this research project to comparatively study the hypothalamus


versus the hippocampus and OB in the context of aging. For our approach, we resorted to DNA methylation epigenetic analysis, given the close relationships between aging and DNA methylation


established in research19,20,21,22,23,24,25,26,27,28,29,30,31,32,33. We were inspired to employ this approach, also because we aimed to explore if age-related DNA methylation in the


hypothalamus could be informative for developing hypothalamic peptide and/or endocrine approaches to address aging-related diseases such as AD. Hence, we carried out this project and


obtained multifaceted data supporting the hypothesis that the hypothalamus is important for modeling age-related DNA methylation epigenetics and developing a related hypothalamic strategy


based on the OXT-GnRH combinational therapy to combat AD. RESULTS AGE-RELATED DNA METHYLATION IN THE HYPOTHALAMUS VS. HIPPOCAMPUS AND OB To profile age-related hypothalamic DNA methylation


in standard C57BL/6 mice, we studied the hypothalamus which was compared to two other limbic system components: the hippocampus and OB. We employed bisulfite oligonucleotide-captured


sequencing (BOCS), a technique established for profiling CG-rich genomic regions. This approach covers all annotated promoters, CG islands, CG shore regions (±2 kb from each CG island), and


CG shelf regions (±2 kb from each shore and ±4 kb from each CG island)34,35. The hypothalamus, hippocampus, and OB were isolated from standard male C57BL/6 mice at the age of 2 months vs. 12


months, representing young and middle age, respectively. Studying the transition from young age to middle age can help identify early changes that could be more causally significant for


subsequent aging-related diseases. All data from this BOCS method were confirmed to meet the standard through quality check (Suppl. Figure 1 ̶ 3). Then, we computed the average methylation


levels of CG and CH sites (H stands for A, T, or C) for each tissue. As shown in Fig. 1A and B, it immediately gained our attention that compared to the hippocampus and OB, DNA methylation


levels were distinctly highest in the hypothalamus of young mice, but the difference tended to be less distinct in middle-aged mice. These overall relationships were also seen in many


individual chromosomes, as demonstrated in chromosome-wise analysis of these brain regions of young mice (Fig. 1C, D) and middle-aged mice (Fig. 1E, F). Taken together, apparently due to


different dynamics of CG methylation and CH demethylation, the pattern of DNA methylation in the hypothalamus distinctly differs from the patterns in the other two brain regions at young


age, but this difference declines when approaching middle age. REGION-RELATED DNA DIFFERENTIAL METHYLATION AT YOUNG AGE AND THE CHANGES BY MIDDLE AGE We then computed age-related


differentially methylated cytosines (DMCs) in each brain region between young and middle age but found only a small number of sites with statistical significance. We also examined how


differential methylation between brain regions could change during the transition to middle age. We found that DMCs between the hypothalamus and the other two brain regions were more heavily


attributed to hypermethylation than to hypomethylation; in contrast, DMCs between the hippocampus and OB were similarly attributed to hypermethylation and hypomethylation. These findings


are summarized in the Volcano plot (Fig. 2) and detailed with chromosome-wide analysis in the Manhattan plot (Suppl. Figure 4, Suppl. Dataset 1). By comparing the two age groups, we found


that DMCs between the hypothalamus and either of the other two brain regions were more than 60% lost by middle age; in contrast, DMCs between the hippocampus and OB in young mice were only


about 40% lost by middle age. Certainly, cellular heterogeneity among different brain regions can importantly account for DMCs between regions, but we expect that composition of major cell


types in a given tissue remains stable by middle age. Thus, we adopted a deconvolution method to test this point, as this approach has been established in the literature to computationally


analyze cell compositions in tissue samples36,37,38,39,40. Our deconvolution analysis led to the projection of 7 major cell types in each brain region (Suppl. Fig. 5); the potential


identities of these major cell types are only suggested, based on previous single-cell analysis of these brain regions in the literature41,42,43. Our analysis supports the point that middle


age does not significantly change the composition of these major cell types. Hence, while we do not exclude a contribution from smaller populations or subtypes, major cell types do not seem


to be a driving factor for changes in region-related differential methylation among these brain tissues as animals approach middle age. AGE-RELATED DIFFERENTIAL METHYLATED REGIONS IN THE


HYPOTHALAMUS, HIPPOCAMPUS AND OB Apart from DMC analysis, we also carried out differential methylation regions (DMRs) analysis, which span lengths of DNA sequences and thus can reveal DNA


methylation information more globally than locally. We analyzed age-dependent DMRs according to DNA length ranging from 300 to 1000 base pairs (bps). While the analysis yielded variable


numbers of DMRs due to changes in DNA lengths, three brain regions consistently exhibited similar patterns, as reflected by an analysis with DNA length of 500 bps (Fig. 3A, Suppl. Dataset 


2). Remarkably, age-dependent DMRs in the hypothalamus exhibited a more balanced distribution between hypermethylation and hypomethylation, in sharp contrast to the unbalanced patterns in


the other two brain regions, where hypermethylation was stronger than hypomethylation (Fig. 3A). Also, as shown in Fig. 3B, chromosome-wide analysis further confirmed a balanced number of


hypothalamic DMRs between hypermethylation and hypomethylation; in contrast, DMRs in the hippocampus and OB were attributed to more hypermethylation than hypomethylation. Subsequently, we


explored the genes involved in age-related DMRs in these brain regions. We identified a pool of genes impacted across the hypothalamus, hippocampus, and OB, constituting approximately 12%,


9%, and 7% of the total affected genes in these brain regions, respectively. Overall, we identified 173 genes mapped onto DMRs due to aging across all three brain regions (Fig. 3C). Among


these 173 genes, 94, 60, and 34 genes showed more hypomethylation than hypermethylation in the hypothalamus, hippocampus, and OB, respectively, indicating a bias towards hypomethylation in


the hypothalamus compared to the other two brain regions. Subsequently, we conducted pathway analysis based on these 173 shared genes. As demonstrated in Fig. 3D, many of these genes were


associated with hypothalamic functions, such as circadian rhythm, reproductive function, endocrine gland hormones, feeding, metabolism, cardiovascular function, and immunity, suggesting that


DNA methylation changes related to hypothalamic function could be shared with other brain regions. PATHWAY ANALYSIS PER AGE-RELATED DMRS IN THE HYPOTHALAMUS, OB, AND HIPPOCAMPUS We


performed further analysis focusing on individual brain regions to explore the specific molecular pathways affected by age-related changes in DNA methylation. Our investigation involved


subsequent mapping of the genes associated with DMRs to a manually curated database of mouse cell signaling and metabolic pathways. Our findings, illustrated in Fig. 4A–C and Suppl. Dataset 


3, highlighted the pathways influenced by age-related differential DNA methylation, characterized by the number of genes and their collective significance. Specifically, the pathways


identified for the hypothalamus included many components that regulate circadian rhythms and the hypothalamic reproductive peptides OXT and GnRH network (Fig. 4A). Of interest, the pathways


identified for the hippocampus and OB were also found to be significantly related to hypothalamic regulatory functions, for example, the circadian entrainment pathway, reproductive OXT


pathway, cortisol synthesis and secretion pathway, Cushing syndrome pathway, and aldosterone synthesis and secretion pathway for the hippocampus (Fig. 4B) and the circadian entrainment


pathway, OXT signaling pathway, and GnRH secretion pathway for OB (Fig. 4C). Most notably, based on two different computational approaches in Figs. 3 and 4, reproductive neuropeptide OXT and


GnRH pathways repeatedly emerged not only in the hypothalamus but also in the hippocampus and OB. In this context, we analyzed how OXT signaling could be related to GnRH signaling using a


protein-protein interaction program, showing a good number of co-components in OXT and GnRH signaling pathways (Suppl. Figure 6). Utilizing BOCS metadata, we investigated how aging affects


the methylation statuses of genes encoding OXT signaling and GnRH signaling components. As laid out in Fig. 4D, by middle age, many components in OXT signaling and GnRH signaling pathways


show age-dependent changes in DNA methylation status in the hypothalamus, hippocampus, and OB. Interestingly, in each brain region, a specific set of OXT and GnRH signaling components was


differentially methylated when comparing young and middle-age samples. Hence, aging is associated with diverse DNA methylation changes in the genes encoding molecular components of OXT and


GnRH signaling, with these variations being brain region-specific. DNA METHYLATION AND TRANSCRIPTION OF OXT AND GNRH GENES IN AGING OR AD-LIKE MODEL Our earlier work separately related GnRH


and OXT to systemic aging or physiology15,16,17,18, altogether suggesting that these peptides might have a close relationship in influencing brain health in conditions such as aging, an idea


which is validated through the DMR analysis presented above. Hence, we decided to directly examine the methylation statuses of _Oxt_ and _Gnrh1_ genes between young and middle-aged mice.


Since the hypothalamus exclusively produces both peptides during adulthood, we focused on analyzing DNA methylation data from the hypothalamic samples. We found that a list of cytosines in


gene body and promoter region of _Oxt_ became hypomethylated, while a smaller list of cytosines became hypermethylated in the hypothalamus by middle age (Fig. 5A, Suppl. Figure 7A). There


was also a list of cytosines in _Gnrh1_ gene body and promoter, which underwent hypomethylation or hypermethylation by middle age (Fig. 5B, Suppl. Figure 7B). Subsequently, we examined the


mRNA levels of _Oxt_ and _Gnrh1_ in the hypothalamus of mice at young age (2 months) compared to middle age (15 months). Through qRT-PCR method, we obtained data showing that mRNA levels of


_Oxt_ and _Gnrh1_ both dramatically dropped in the hypothalamus during the transition from young to middle age (Fig. 5C). Given our interest in aging-related disease, we employed 5xFAD mice,


an AD-like model associated with aging, to assess if similar changes might occur in the hypothalamus prior to the onset of AD-like phenotype. Indeed, the transcription levels of _Oxt_ and


_Gnrh1_ were also evidently downregulated in the hypothalamus of 5xFAD mice compared to wildtype controls (Fig. 5D). These results lend support to our prediction that declines in OXT and


GnRH pathways are co-involved in AD-like conditions which are associated with aging. DNA METHYLATION AND TRANSCRIPTION OF ADCY FAMILY MEMBERS IN AGING OR AD-LIKE MODEL In addition to _Oxt_


and _Gnrh1_, we noted that our age-related DMR analysis across the hypothalamus, hippocampus, and OB shown in Fig. 4 contained several adenylate cyclases (Adcy), a family of proteins that


co-mediate OXT and GnRH signaling (Suppl. Fig. 6). By modulating cAMP levels, Adcy members link OXT and GnRH stimulation to downstream protein kinases that regulate various events ranging


from gene expression and neurotransmitter release to synaptic plasticity. Abnormal _Adcy_ expression is associated with many neurological disorders, including AD and depressive


disorders44,45. For instance, loss of _Adcy5_ results in Parkinson’s disease-like disorders46, and _Adcy7_ is linked to familial major depression in both mice and humans47,48. Hence, our


methylation analysis further narrowed down to the promoter regions of _Adcy_ members, given the important role of promoters in gene expression, focusing on the hippocampus because of its


critical role in cognitive regulation. We found that total methylation levels in the promoter regions of several _Adcy_ genes were significantly lower in middle-aged mice compared to young


mice, while _Adcy_ genes often underwent hypomethylation in the proximal promoter regions by middle age (Fig. 6A). In this context, we examined hippocampal mRNA levels and found that middle


age was associated with significant downregulation across many _Adcy_ genes (Fig. 6B). Also, we examined the hippocampus of 5xFAD model and observed that hippocampal mRNA levels of several


_Adcy_ members were lower in this AD-like model compared to littermate controls (Fig. 6C). Taken together, transcription of _Oxt_ and _Gnrh1_ in the hypothalamus, as well as transcription of


_Adcy_ members in the hippocampus, are commonly defective in both the aging and the 5xFAD model. THERAPEUTIC SIGNIFICANCE OF OXT-GNRH COMBINATIONAL TREATMENT IN AD MOUSE MODEL While this


work began with DNA methylation analysis based on aging model, our analysis led to identification of OXT and GnRH pathways, which we confirmed at the gene transcriptional level to be


significantly altered in an AD-like model which is associated with aging. In the literature, a few studies recently suggested an effect of OXT in attenuating amyloid β (Aβ) deposition or


cognitive impairment in ddY or APP/PS1 mice or Sprague Dawley rats49,50,51,52,53,54. Since these studies were based on experimental conditions with relatively mild cognitive disorders, the


effects of OXT in treating AD still remain to be defined. Regarding GnRH, researchers have studied its potential in treating diseases such as cancers, and recently, it was shown to have an


effect in improving cognition in Down syndrome and dementia55,56,57, but the effect of GnRH on AD is unclear and it remains completely unknown if OXT and GnRH might be used in combination to


synergistically and effectively combat against AD. Hence, we studied a 9-month-old 5xFAD mouse model with strong manifestation of AD-like phenotypes, focusing to compare OXT-GnRH


combinational treatment vs. individual peptide treatment. We employed nasal administration, a method which has often been used in research to deliver peptides into the brain for treating


neurological diseases58,59,60,61,62,63,64. To help discern the effects of combinational vs. individual treatment, OXT and GnRH were administered nasally at relatively low doses (50 ng and 5 


ng, respectively) for a relatively short treatment duration (2 months). After the completion of therapy, animals were assessed with a battery of non-invasive neurobehavioral assays,


including an open field, grip strength, y-maze, novel object recognition, social interaction, and Morris Water Maze for various aspects of physical and cognitive functions. As demonstrated


in Fig. 7, OXT-GnRH combinational treatment led to very robust and strong effects against various neurological disorders in this AD-like model, while the effects from single peptide


treatment were modest or non-significant. At the end, we assessed how OXT-GnRH therapy could affect Aβ levels in this model. Indeed, 5×FAD model had a very large amount of Aβ plaques


throughout various brain regions, represented by hippocampal CA1, CA3 and dentate gyrus (DG), entorhinal cortex, and mediobasal hypothalamus. In agreeing with the physiological effects,


OXT-GnRH combinational treatment led to a nearly complete reversal of Aβ deposition (Fig. 8). Compared to the combinational treatment, single peptide treatments were much less effective,


except that GnRH treatment was also sufficient to reduce Aβ plaques in the mediobasal hypothalamus, while Aβ plaques in fornix bundle in this region were not amended by any treatment. Taken


together, compared to single peptide treatment, OXT-GnRH combinational therapy increased the sensitivity and effectiveness in treating neurological disorders and reducing Aβ plaques in male


5×FAD model. Finally, given the observed transcriptional changes of _Adcy_ members in 5xFAD model (Fig. 6), we additionally assessed how OXT-GnRH treatment, compared to individual peptide


treatment, might affect promoter methylation of these _Adcy_ genes. Thus, we utilized the Next-Generation Sequencing-based Bisulfite Sequencing PCR (NGS-BSP) method to sequence the promoter


subregions of candidate _Adcy_ genes, focusing on subregions between CG island and encoding sequence. We performed target sequencing on hippocampal tissues obtained from the 5xFAD mice


following combinational versus individual treatment with OXT and GnRH, as described in Fig. 7. Although either treatment did not cause significant changes in average methylation levels of


the promoter subregions which we sequenced, our DMC analysis did lead to identification of several short sequences in which cytosine methylation levels were altered differently by OXT-GnRH


treatment compared to single peptide treatment (Suppl. Fig. 8). Admittedly, this research is still limited, but it might point to a future direction for studying loci effects of OXT-GnRH


treatment on gene promoter activity and if it could significantly affect the transcription of key genes in contributing to the combinational therapy against AD-like pathologies. DISCUSSION


In this project, we computationally analyzed age-related DNA methylation in the hypothalamus compared to two other limbic system components in a standard mouse model. This analysis led to


the discovery that the hypothalamus is characterized by distinctively high-level DNA methylation at young age and the distinct dynamics of DNA methylation and demethylation when approaching


middle age. Thus, compared to the other two limbic system regions, the hypothalamus contains more DNA methylation information during young age, contributing to its unique pattern of DNA


methylation prior to middle age. During the course of approaching middle age, this distinction of DNA methylation between the hypothalamus and the other limbic system regions becomes to be


declined. Thus, while aligning with the ‘hypothalamic control of aging’ paradigm which we previously proposed12,14,15, we further speculate that hypothalamic regulation of mammalian aging


could be causally related to loss of epigenetic information from DNA methylation in the genomes of the hypothalamus. In this context, more brain regions should be compared to the


hypothalamus in light of age-related DNA methylation. We also surprisingly found that not only the hypothalamus but also other limbic system regions commonly involve age-related DNA


methylation of genes related to various hypothalamic endocrine pathways and notably OXT and GnRH pathways. This finding suggests the importance of the hypothalamus in age-related brain


regional DNA methylation, warranting future research. Furthermore, the identification of OXT and GnRH pathways in this computational analysis lends support for investigating whether these


neuropeptides could be designed to target aging-related diseases. Hence, using an AD-like condition as an important example of aging-related diseases, we focused intensively on hypothalamic


neuropeptide OXT and GnRH pathways for the potential therapeutic development. Following the observation that multiple components in OXT and GnRH pathways become declined in relevant brain


regions at the gene transcriptional level in an AD-like mouse model, we were inspired to evaluate the effects of these two neuropeptides in combination versus individually for treating


AD-like pathologies in this mouse model. This effort led to a major finding, that is, OXT-GnRH combinational therapy had a robust effect against the severe symptoms in an aged 5×FAD mouse


model, showing significantly greater efficacy than individual peptide treatments. This finding can suggest a clinical strategy of using OXT-GnRH combinational therapy to target AD, which


would call for clinical research down the road. In the literature, OXT has recently gained some attention mostly for reducing Aβ deposition49,50,51,52,53,54, while it remains unknown if GnRH


could be used to treat AD. It is likely that individual peptides, especially when dose and treatment conditions are stronger, can each have a better effect on AD-like disorders, but


strategy of developing combinational treatments should have advantages, for instance, low dose of each peptide helps minimize off-target effects, and some therapeutic effects are expected to


require interactions and synergy of the two neuropeptide pathways which might not be achievable by individual peptides. Our study still has limitations, mostly due to practical constraints


during developing this project. For the computational study, we have not compared the hypothalamus with other limbic system components or other brain regions; to do so will be valuable to


solidify the model on hypothalamic DNA methylation modeling of aging. Technically, we recognize a limitation that we employed 3 replicates of animals per group in our computational assays


although it was often similarly used in the literature65,66,67, hence, to include more samples will likely lead to more information. Moreover, while our computational work was based on an


aging model, it would be more informative to include samples from aging-related diseases such as AD. For the therapeutic study, while our work was to initially establish the proof of


principle for using OXT-GnRH combination to target AD-like pathologies, future research is still needed to optimize dosage and duration, to assess additional ages and AD conditions, and to


profile any off-target effects. In particular, we acknowledge a limitation due to the absence of studies on female model, which needs future research to address. Last but not least, while


our therapeutics was based on an AD-like model as an example of aging-related disease, it will be very valuable to assess if this strategy could apply to other aging-related diseases, for


example, Parkinson’s disease. METHODS This study was conducted in compliance with all relevant ethical regulations. The animal research protocol was reviewed and approved by the


Institutional Animal Care and Use Committee of the Albert Einstein College of Medicine (Protocols #00001111, #00001385, #00001397, #00001398, #00001399 and their previously approved


versions). ANIMAL MODELS C57BL/6 mice and 5×FAD mice were obtained from Jackson Laboratory and housed under standard conditions in a 12-h light/12-h dark cycle with free access to food and


water. All mice in this study were male. All mice were kept under standard, infection-free housing with 3 to 5 mice per cage. Pathogen-free quality of mouse colonies was ensured through


quarterly serology, quarterly histopathologic exams and daily veterinarian monitoring of animal health and care. All mice were fed a standard chow from Lab Diet. Perfusion-induced euthanasia


was used for tissue sampling and carbon dioxide inhalation euthanasia was used for all other mice which were generated during the study according to the IACUC approved protocols. BISULFITE


OLIGONUCLEOTIDE-CAPTURED SEQUENCING (BOCS) The hypothalamus, hippocampus, and OB were obtained from standard, chow-fed male C57BL/6 mice at 2 vs. 12 months of age (_n_ = 3 mice per group).


DNA was extracted from these tissues using a Qiagen kit (Cat# 69506). The extracted DNA samples were then sheared into fragments of 150–200 base pairs using sonication, and the fragment


sizes were verified using the Agilent TapeStation with D1000 ScreenTape. The fragmented DNA underwent library preparation using the SureSelectXT Methyl-Seq Library Prep Kit, following the


manufacturer’s instructions. The generated libraries were subsequently hybridized with the SureSelectXT Mouse Methyl-Seq capture set for 16 h at 65 °C, specifically targeting all CG regions


including all annotated promoters, CG islands and shore regions (±2 kb from islands) as well as shelf regions (±2 kb from shores and ±4 kb from islands). The hybridized libraries were


captured using DynaBeads MyOne Streptavidin T1 magnetic beads. The captured libraries were eluted from magnetic beads using 0.1 M NaOH. Unmethylated cytosine residues in the captured DNA


libraries were then converted to uracil through bisulfite conversion using the EZ DNA Methylation-Gold Kit from Zymo Research. Following bisulfite conversion, DNA was desulfonated and


amplified via PCR. The amplified libraries were purified using AMPure XP beads. The purified libraries were indexed, allowing for the multiplexing of samples in a single sequencing run.


Quantification of DNA libraries was performed using the TapeStation with D1000 ScreenTape. The libraries were normalized to a concentration of 4 nM, pooled together, and further diluted to a


final concentration of 12 pM. The DNA libraries were sequenced on an Illumina MiSeq PE75 for quality control and sequenced on NextSeq500 High PE75 to obtain DNA methylation profiles. DNA


METHYLATION ANALYSIS FASTQ files were accessed from bisulfite sequencing for quality control with FastQC and paired-end reads were trimmed for quality with Illumina’s BaseSpace sequence hub


having MethylSeq v2.0.0. Trimming involved removing 3 nucleotides at the 5′ end and 4 nucleotides at the 3′ end, as well as adapter removal using FASTQ Toolkit v2.2. Only reads with a


Q-score >30 were used for mapping, reads which did not meet criteria after trimming were discarded. Alignment of trimmed bisulfite converted sequences was carried out using Bismark


Bisulfite Mapper v0.14.4/Bowtie 2 v2.2.2 against the mouse reference genome (GRCm38/mm10)68,69. Each sample yielded over 25 million aligned reads, resulting in an average target sequence


coverage ranging from 18 to 20×. The aligned BAM files were processed using SAMtools v1.270. After read mapping, methylation calling was carried out using the Bismark methylation extractor.


The percentage of methylation was determined by calculating the ratio of methylated cytosines to the total number of cytosines covered in the genome for each specific cytosine position.


Differential methylation analysis was primarily conducted in R v4.1.2, using MethylKit v1.20.071. Differentially methylated cytosines were determined from sites passing coverage criteria of


minimum 10. Logistic regression was employed to determine differentially methylated sites between tissues, and p-values were adjusted using a false-discovery rate (q-value). Sites with


q-value < 0.05 and methylation difference of ≥25% were considered statistically significant. For computing DMRs, criteria were set at a length of 500 bp showing >10% average


methylation difference. Gene annotation files for the mouse (GRCm38) were downloaded from Gencode and mapped with DMRs using BEDTools v2.30.071,72. Deconvolution analysis, a method for


analyzing cellular heterogeneity based on methylation sequencing data36,73,74,75, was used to computationally assess tissue heterogeneity of cell populations. The computation was based on


using DXM (Deconvolution of Subpopulations Existing in Methylation Data), a reference-free approach offering versatility across various Bisulfite Sequencing (BS) methodologies39 which was


established in the literature to accurately infer cell-type proportions40. DXM’s initial module “dxm_estimateFracs” was used within the Python environment. Enrichr was used to perform KEGG


pathway analysis, allowing the identification and enrichment analysis of genes within specific biological pathways76. Cytoscape, in conjunction with the String module, with confidence


>0.4 as the cut-off criteria, was employed to construct protein-protein interaction network, visualization and analysis of interconnections between pathway components77,78. Data were


processed and visualized in Python, and computational analyses were conducted using custom scripts or published tools in UNIX, R and Python environments. ANIMAL TREATMENT AND BEHAVIORAL


ASSAYS Male 5xFAD mice under standard housing and feeding were randomly assigned into various treatment groups to receive 50 ng OXT (Cat # 04375-1000IU, Sigma Aldrich) and 5 ng GnRH (Cat #


PEP-168, Invitrogen) treatment through daily nasal administration. Following 2-month treatment, animals were sequentially examined with a panel of non-invasive behavioral assays with


adequate recovery time between different assays. All behavioral tests were performed in an isolated and designated mouse behavioral room. An anymaze video-tracking system (version 4.99 m,


Stoelting) connected with a digital camera and computer was used to record the activities of mice during behavioral procedures. Grip test: Each mouse was positioned on a square grid with a


1-cm mesh, which was then inverted 30 cm above a protective pad. The mouse was then allowed to hang by its paws for a specified period, and the duration for which the mouse remained


suspended was recorded. Three repeats were performed for each animal with 30 min rest time between trials. Open field test: A mouse was placed in the lower right corner of a spacious, empty


white plastic chamber (40 cm length × 40 width and × 40 cm height) and was allowed to freely move in the chamber while movement and behavior were recorded by the camera for 10 min. Y-maze


test: A mouse was placed in the center of Y-maze apparatus and allowed to freely explore for 10 min. The time spent in each arm and the number of arm entries were recorded and analyzed. An


entry was considered a success when all four limbs entered an arm. Novel object recognition: A mouse was allowed to freely explore an open-field box (40 cm in length, 40 cm in width, and 40 


cm in height) for 10 min prior to experimental sessions. During the familiarization session, the mouse was then allowed to freely explore two similar objects for 10 min. During the test


session, one of the two objects was replaced by a novel object for 10 min. The amount of time that the mouse spent exploring each object was recorded. A preference index was calculated using


the ratio of the amount of time exploring each object over the total time exploring both objects. Morris water maze (MWM): The test was performed using a round water tank (90 cm in


diameter) containing water at temperature of 22 to 23 °C and a non-toxic paint which was added to make opaque and white background. A circular, background color-matched platform with a


diameter of 10 cm was placed 25 cm from the wall of the tank and about 1 cm below the water surface which was thus invisible. Animals were trained for 5 days, 4 training sessions each day,


and the beginning positions for each training day were randomly chosen. At each training session, mice were placed in a beginning position in the maze and were trained to find the platform


within 60 s. Latency to reach the platform of each trial was recorded. On the next day following 5-day training sessions, probe trials were performed by removing the platform and the animal


was allowed to swim for 60 s and measured for swimming speed and the latency that the mouse swam to cross the location where the platform was previously located. Sociality: Social


interaction was tested in a gray 3-chamber neutral box cage (60 cm in length, 40 cm in width, and 22 cm in height). For adaptation phase, animals were allowed to explore freely for 10 min in


the neutral cage for habituating the testing conditions. For social affiliation phase, a new mouse (stranger) in a wire containment cup was placed in a side chamber. The subject mouse was


allowed free access to explore each of three chambers for 10 min. For preference testing phase, a second new mouse (new stranger) in a wire containment cup was placed in the opposite side


chamber. The subject mouse was allowed to freely explore each of three chambers for 10 min. The time spent in social interaction by sniffing was recorded. BRAIN SECTIONS AND IMMUNOSTAINING


The mice from the above therapeutic experiment were used for brain Aβ immunostaining (subgroup of brain samples from these mice used for DNA promoter target sequencing). To obtain brain


samples, mice were anesthetized with 3–5% isoflurane inhalation and were transcardially perfused with saline over 5 min before brains were collected. Brain hemispheres were post-fixed with


4% PFA and then infiltrated with 20–30% sucrose for immunostaining. Brain sections were generated with a thickness of 20 μm using a cryostat. To facilitate Aβ immunostaining, sections were


treated with 88% formic acid to expose the antigen, followed by blocking with a buffer solution containing 5% BSA, 5% goat serum, and 0.1% Triton X-100 in PBS. Subsequently, brain sections


were incubated overnight at 4 °C with mouse anti-Aβ primary antibody (anti-β-Αmyloid, BioLegend, #803003, 1:500) in a blocking buffer. Technical controls included appropriate species-matched


naive IgGs. Following three washes, the sections were incubated with secondary antibody (Goat anti-mouse IgG antibody Alexa Fluor 555, Thermo Fisher, #A21422, 1:500), and after three


washes, sections were covered with a mounting medium containing DAPI for visualizing the nuclei of cells in the sections. IMAGE CAPTURE AND QUANTIFICATION Brain sections were imaged for a


target region using the LAS X software-equipped Leica Stellaris 8 confocal microscope. The same confocal setting and parameters were used for all samples in this study. Confocal scanning was


set at 600 Hz scan speed, 4 lines average, and 2 frame average. 20×/0.75 dry objective was used for imaging hippocampal subregions, and 10×/0.40 dry objective used for imaging other brain


regions. Aβ plaque signals were imaged with an ALEXA channel at excitation wavelength of 553 nm and emission light collected with a wavelength of 558 to 730 nm. DAPI staining signal was


imaged at excitation wavelength of 405 nm and emission light collected with a wavelength of 430 to 558 nm. The pinhole size was set at 56.7 μm for both channels. Images were exported as


scaled viewer tiff files with lossless compression. ImageJ 1.54 f was used to quantitate image signal of interest. The same settings of ImageJ were used to process all images in this study.


RGB images were converted into 8-bit grayscale format for each channel by ImageJ, and the entire region of interest (ROI) was delineated according to the anatomic structure. Aβ plaques in an


ROI were subsequently identified using a threshold signal of 122 to 255 scale, and the plaque density of a target brain region was calculated as Aβ signal per area of ROI. QUANTITATIVE


RT-PCR The hypothalamus and hippocampus samples were obtained from standard male C57BL/6 mice at the age of 2 vs. 15 months and from male 5xFAD mice and littermate male WT controls at the


age of 8 months for extraction of total RNA using TRIzol reagent (Invitrogen), followed by reverse transcription using High-Capacity RNA to cDNA kit (Thermo Fisher) according to the


manufacturer’s instructions. Quantitative real-time PCR was conducted using Power SYBR Green PCR Master Mix (Thermo Fisher) with specific primer sets (see below). β-actin was used as a


reference and changes were calculated using 2-ΔΔCt method. _Oxt_: 5ʹ-GCTGCCAGGAGGAGAACTAC-3ʹ; 5ʹ-GGCAGCCATCTGCAAGAGAA-3ʹ _Gnrh1_: 5ʹ-GCATTCTACTGCTGACTGTGTGTT-3ʹ; 5ʹ-GTTCTGCCATTTGATCCACCT-3ʹ


_Adcy3_: 5ʹ-TCTTTGACTGCTACGTGGTAGT-3ʹ; 5ʹ-GGCCCGTGAAAAGTTCAGG-3ʹ _Adcy5_: 5ʹ-AAGATCCTCGGGGATTGTTACT-3ʹ; 5ʹ-CTCCCGGACCAACGAGATG-3ʹ _Adcy7_: 5ʹ-AAGGGGCGCTACTTCCTAAAT-3ʹ;


5ʹ-GTGTCTGCGGAGATCCTCA-3ʹ _Adcy9_: 5ʹ-CAACAGCGTGAGGGTCAAGAT-3ʹ; 5ʹ-CATGGAGTCGAATTTGGGGTC-3ʹ _β-actin_: 5ʹ-CCTCTATGCCAACACAGTGC-3ʹ; 5ʹ-GCTAGGAGCCAGAGCAGTAA-3ʹ STATISTICS AND REPRODUCIBILITY


Sample sizes were designed considering the relevant literature for physiological, biochemical and histological experiments12,14,15,79,80, relevant literature for DNA bisulfite


sequencing65,66,67, and relevant literature for target bisulfite sequencing67,81. Neurobehavioral tests were performed making group information blind to experimental performers when


collecting the data. All physiological and histological results represented repeated observations independently and through complimentary approaches. All data with biological replicates were


presented as mean ± S.E.M. The number of biological replicates and applied statistical methods are detailed in figure legends. Data were analyzed for parametric or non-parametric


distributions with statistical tools such as Shapiro-Wilk test, D’Agostino-Pearson test, Anderson-Darling test, and Kolmogorov-Smirnov test. Data that followed parametric distribution were


analyzed using ANOVA with Tukey’s multiple comparisons when involving more than two groups and two-tailed unpaired Student’s t-test when involving only two groups. Data that did not follow


parametric distribution were analyzed including Kruskal-Wallis test and Dunn’s multiple comparisons when having more than two groups and two-tailed Mann-Whitney test when having only two


groups. Additional statistics included logistic regression and Fisher’s Exact test contained in applied computational programs. Statistical significance of all data was determined at _p_ 


< 0.05. For computing differential methylation sites and regions, logistic regression-based modeling was applied in R-based MethylKit v1.20.0, with a false discovery rate set at _q_ < 


0.05. REPORTING SUMMARY Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article. DATA AVAILABILITY BOCS datasets for all


biological samples generated in this study are available in the NCBI Gene Expression Omnibus (GEO) repository with accession number GSE276875. Target promoter subregional data and raw


immunofluorescence images are deposited in publicly accessible platform Figshare (https://figshare.com/s/c2ed30d10acfa8a43f56). Source data are provided with this paper. There is no


restriction on data availability in this manuscript, and research materials developed from this study are freely available to research upon request. Source data are provided with this paper.


CODE AVAILABILITY Code/scripts for computational analysis are deposited in the publicly accessible GitHub repository


(https://github.com/DongshengCai/Brain-subregional-DNA-methylation-aging). REFERENCES * Shohayeb, B., Diab, M., Ahmed, M. & Ng, D. C. H. Factors that influence adult neurogenesis as


potential therapy. _Transl. Neurodegener._ 7, 4 (2018). Article  PubMed  PubMed Central  Google Scholar  * Culig, L., Chu, X. & Bohr, V. A. Neurogenesis in aging and age-related


neurodegenerative diseases. _Ageing Res. Rev._ 78, 101636 (2022). Article  CAS  PubMed  PubMed Central  Google Scholar  * Dean, D. C. et al. Brain differences in infants at differential


genetic risk for late-onset Alzheimer disease: a cross-sectional imaging study. _Jama Neurol._ 71, 11–22 (2014). Article  PubMed  PubMed Central  Google Scholar  * Fu, H., Hardy, J. &


Duff, K. E. Selective vulnerability in neurodegenerative diseases. _Nat. Neurosci._ 21, 1350–1358 (2018). Article  CAS  PubMed  PubMed Central  Google Scholar  * van Praag, H. et al.


Functional neurogenesis in the adult hippocampus. _Nature_ 415, 1030–1034 (2002). Article  ADS  PubMed  PubMed Central  Google Scholar  * Kuhn, H. G., Dickinson-Anson, H. & Gage, F. H.


Neurogenesis in the dentate gyrus of the adult rat: age-related decrease of neuronal progenitor proliferation. _J. Neurosci._ 16, 2027–2033 (1996). Article  CAS  PubMed  PubMed Central 


Google Scholar  * Lois, C. & Alvarez-Buylla, A. Proliferating subventricular zone cells in the adult mammalian forebrain can differentiate into neurons and glia. _Proc. Natl. Acad. Sci.


USA_ 90, 2074–2077 (1993). Article  ADS  CAS  PubMed  PubMed Central  Google Scholar  * Alvarez-Buylla, A. & Lois, C. Neuronal stem cells in the brain of adult vertebrates. _Stem Cells


(Dayt. Ohio)_ 13, 263–272 (1995). Article  CAS  Google Scholar  * Doetsch, F., Caillé, I., Lim, D. A., García-Verdugo, J. M. & Alvarez-Buylla, A. Subventricular zone astrocytes are


neural stem cells in the adult mammalian brain. _Cell_ 97, 703–716 (1999). Article  CAS  PubMed  Google Scholar  * Alvarez-Buylla, A. & Garcia-Verdugo, J. M. Neurogenesis in adult


subventricular zone. _J. Neurosci._ 22, 629–634 (2002). Article  CAS  PubMed  Google Scholar  * Xiao, Y. Z. et al. Reducing hypothalamic stem cell senescence protects against


aging-associated physiological decline. _Cell Metab._ 31, 534–548.e535 (2020). Article  CAS  PubMed  Google Scholar  * Zhang, Y. L. et al. Hypothalamic stem cells control ageing speed partly


through exosomal miRNAs. _Nature_ 548, 52–57 (2017). Article  ADS  CAS  PubMed  Google Scholar  * Li, J. X., Tang, Y. Z. & Cai, D. S. IKK beta/NF-kappa B disrupts adult hypothalamic


neural stem cells to mediate a neurodegenerative mechanism of dietary obesity and pre-diabetes. _Nat. Cell Biol._ 14, 999–1012 (2012). Article  CAS  PubMed  Google Scholar  * Wang, Z., Wu,


W., Kim, M. S. & Cai, D. GnRH pulse frequency and irregularity play a role in male aging. _Nat. Aging_ 1, 904–918 (2021). Article  PubMed  Google Scholar  * Zhang, G. et al. Hypothalamic


programming of systemic ageing involving IKK-beta, NF-kappaB and GnRH. _Nature_ 497, 211–216 (2013). Article  ADS  CAS  PubMed  Google Scholar  * Tang, Y. & Cai, D. Hypothalamic


inflammation and GnRH in aging development. _Cell Cycle_ 12, 2711–2712 (2013). Article  CAS  PubMed  Google Scholar  * Zhang, G. & Cai, D. Circadian intervention of obesity development


via resting-stage feeding manipulation or oxytocin treatment. _Am. J. Physiol. Endocrinol. Metab._ 301, E1004–E1012 (2011). Article  CAS  PubMed  Google Scholar  * Zhang, G. et al.


Neuropeptide exocytosis involving synaptotagmin-4 and oxytocin in hypothalamic programming of body weight and energy balance. _Neuron_ 69, 523–535 (2011). Article  CAS  PubMed  PubMed


Central  Google Scholar  * Horvath, S. Erratum to: DNA methylation age of human tissues and cell types. _Genome Biol._ 16, 96 (2015). Article  PubMed  PubMed Central  Google Scholar  *


Hannum, G. et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. _Mol. Cell_ 49, 359–367 (2013). Article  CAS  PubMed  Google Scholar  * Jones, M. J.,


Goodman, S. J. & Kobor, M. S. DNA methylation and healthy human aging. _Aging Cell_ 14, 924–932 (2015). Article  CAS  PubMed  Google Scholar  * Pal, S. & Tyler, J. K. Epigenetics and


aging. _Sci. Adv._ 2, e1600584 (2016). Article  ADS  PubMed  Google Scholar  * Dor, Y. & Cedar, H. Principles of DNA methylation and their implications for biology and medicine. _Lancet


(Lond., Engl.)_ 392, 777–786 (2018). Article  CAS  Google Scholar  * Hou, Y. et al. Ageing as a risk factor for neurodegenerative disease. _Nat. Rev. Neurol._ 15, 565–581 (2019). Article 


PubMed  Google Scholar  * Unnikrishnan, A. et al. The role of DNA methylation in epigenetics of aging. _Pharmacol. Therapeutics_ 195, 172–185 (2019). Article  CAS  Google Scholar  * Zhang,


W., Qu, J., Liu, G. H. & Belmonte, J. C. I. The ageing epigenome and its rejuvenation. _Nat. Rev. Mol. Cell Biol._ 21, 137–150 (2020). Article  CAS  PubMed  Google Scholar  * Sikora, E.


et al. Cellular senescence in brain aging. _Front Aging Neurosci._ 13, 646924 (2021). Article  CAS  PubMed  Google Scholar  * Rutledge, J., Oh, H. & Wyss-Coray, T. Measuring biological


age using omics data. _Nat. Rev. Genet_ 23, 715–727 (2022). Article  CAS  PubMed  Google Scholar  * Seale, K., Horvath, S., Teschendorff, A., Eynon, N. & Voisin, S. Making sense of the


ageing methylome. _Nat. Rev. Genet_ 23, 585–605 (2022). Article  CAS  PubMed  Google Scholar  * Wang, K. et al. Epigenetic regulation of aging: implications for interventions of aging and


diseases. _Signal Transduct. Target Ther._ 7, 374 (2022). Article  CAS  PubMed  PubMed Central  Google Scholar  * He, X., Memczak, S., Qu, J., Belmonte, J. C. I. & Liu, G. H. Single-cell


omics in ageing: a young and growing field. _Nat. Metab._ 2, 293–302 (2020). Article  PubMed  Google Scholar  * Horvath, S. & Raj, K. DNA methylation-based biomarkers and the epigenetic


clock theory of ageing. _Nat. Rev. Genet._ 19, 371–384 (2018). Article  CAS  PubMed  Google Scholar  * Yang, J. H. et al. Loss of epigenetic information as a cause of mammalian aging.


_Cell_ https://doi.org/10.1016/j.cell.2022.12.027 (2023). Article  PubMed  PubMed Central  Google Scholar  * Masser, D. R. et al. Sexually divergent DNA methylation patterns with hippocampal


aging. _Aging Cell_ 16, 1342–1352 (2017). Article  CAS  PubMed  PubMed Central  Google Scholar  * Masser, D. R. et al. Bisulfite oligonucleotide-capture sequencing for targeted base- and


strand-specific absolute 5-methylcytosine quantitation. _Age (Dordr.)_ 38, 49 (2016). Article  PubMed  Google Scholar  * Song, J. & Kuan, P. F. A systematic assessment of cell type


deconvolution algorithms for DNA methylation data. _Brief Bioinform._ 23, https://doi.org/10.1093/bib/bbac449 (2022). * Rahmani, E. et al. Correcting for cell-type heterogeneity in DNA


methylation: a comprehensive evaluation. _Nat. Methods_ 14, 218–219 (2017). Article  CAS  PubMed  Google Scholar  * Rahmani, E. et al. Cell-type-specific resolution epigenetics without the


need for cell sorting or single-cell biology. _Nat. Commun._ 10, 3417 (2019). Article  ADS  PubMed  PubMed Central  Google Scholar  * Fong, J. et al. Determining subpopulation methylation


profiles from bisulfite sequencing data of heterogeneous samples using DXM. _Nucleic Acids Res._ 49, e93 (2021). Article  CAS  PubMed  PubMed Central  Google Scholar  * Jeong, Y. et al.


Systematic evaluation of cell-type deconvolution pipelines for sequencing-based bulk DNA methylomes. _Brief Bioinform._ 23, https://doi.org/10.1093/bib/bbac248 (2022). * Hajdarovic, K. H. et


al. Single-cell analysis of the aging female mouse hypothalamus. _Nat. Aging_ 2, 662–678 (2022). Article  CAS  PubMed  PubMed Central  Google Scholar  * Yang, Y. T. et al. STAB2: an updated


spatio-temporal cell atlas of the human and mouse brain. _Nucleic Acids Res._ 52, D1033–D1041 (2024). Article  PubMed  Google Scholar  * Tepe, B. et al. Single-cell RNA-Seq of mouse


olfactory bulb reveals cellular heterogeneity and activity-dependent molecular census of adult-born neurons. _Cell Rep._ 25, 2689–2703.e2683 (2018). Article  CAS  PubMed  PubMed Central 


Google Scholar  * Devasani, K. & Yao, Y. Expression and functions of adenylyl cyclases in the CNS. _Fluids Barriers CNS_ 19, 23 (2022). Article  CAS  PubMed  PubMed Central  Google


Scholar  * Mons, N., Segu, L., Nogues, X. & Buhot, M. C. Effects of age and spatial learning on adenylyl cyclase mRNA expression in the mouse hippocampus. _Neurobiol. Aging_ 25,


1095–1106 (2004). Article  CAS  PubMed  Google Scholar  * Iwamoto, T. et al. Motor dysfunction in type 5 adenylyl cyclase-null mice. _J. Biol. Chem._ 278, 16936–16940 (2003). Article  CAS 


PubMed  Google Scholar  * Joeyen-Waldorf, J. et al. Adenylate cyclase 7 is implicated in the biology of depression and modulation of affective neural circuitry. _Biol. Psychiatry_ 71,


627–632 (2012). Article  CAS  PubMed  PubMed Central  Google Scholar  * Hines, L. M. et al. A sex-specific role of type VII adenylyl cyclase in depression. _J. Neurosci._ 26, 12609–12619


(2006). Article  CAS  PubMed  PubMed Central  Google Scholar  * Takahashi, J. et al. Oxytocin reverses Aβ-induced impairment of hippocampal synaptic plasticity in mice. _Biochem Biophys. Res


Commun._ 528, 174–178 (2020). Article  CAS  PubMed  Google Scholar  * Takahashi, J. et al. Intracerebroventricular administration of oxytocin and intranasal administration of the oxytocin


derivative improve β-amyloid peptide (25-35)-induced memory impairment in mice. _Neuropsychopharmacol. Rep._ 42, 492–501 (2022). Article  CAS  PubMed  Google Scholar  * El-Ganainy, S. O. et


al. Intranasal oxytocin attenuates cognitive impairment, β-amyloid burden and tau deposition in female rats with Alzheimer’s disease: interplay of ERK1/2/GSK3β/caspase-3. _Neurochem. Res._


47, 2345–2356 (2022). Article  CAS  PubMed  Google Scholar  * Selles, M. C. et al. Oxytocin attenuates microglial activation and restores social and non-social memory in APP/PS1 Alzheimer


model mice. _iScience_ 26, 106545 (2023). Article  ADS  CAS  PubMed  Google Scholar  * Koulousakis, P. et al. Exogenous oxytocin administration restores memory in female APP/PS1 Mice. _J.


Alzheimers Dis._ 96, 1207–1219 (2023). Article  CAS  PubMed  Google Scholar  * Ye, C. et al. Oxytocin nanogels inhibit innate inflammatory response for early intervention in Alzheimer’s


disease. _ACS Appl Mater. Interfaces_ 14, 21822–21835 (2022). Article  CAS  PubMed  Google Scholar  * Garrido, M. P., Hernandez, A., Vega, M., Araya, E. & Romero, C. Conventional and new


proposals of GnRH therapy for ovarian, breast, and prostatic cancers. _Front Endocrinol. (Lausanne)_ 14, 1143261 (2023). Article  PubMed  Google Scholar  * Sterling, K., Cao, R. & Song,


W. Gonadotropin releasing hormone (GnRH): a hormone therapy boosts cognition in Down syndrome and dementia. _Signal Transduct. Target Ther._ 8, 49 (2023). Article  CAS  PubMed  Google


Scholar  * Manfredi-Lozano, M. et al. GnRH replacement rescues cognition in Down syndrome. _Science_ 377, eabq4515 (2022). Article  CAS  PubMed  Google Scholar  * Patel, D., Patel, B. &


Wairkar, S. Intranasal delivery of biotechnology-based therapeutics. _Drug Discov. Today_ 27, 103371 (2022). Article  CAS  PubMed  Google Scholar  * Akita, T. et al. Involvement of


trigeminal axons in nose-to-brain delivery of glucagon-like peptide-2 derivative. _J. Control Release_ 351, 573–580 (2022). Article  CAS  PubMed  Google Scholar  * Finger, E. et al. Adaptive


crossover designs for assessment of symptomatic treatments targeting behaviour in neurodegenerative disease: a phase 2 clinical trial of intranasal oxytocin for frontotemporal dementia


(FOXY). _Alzheimers Res Ther._ 10, 102 (2018). Article  PubMed  Google Scholar  * Lindenmaier, Z. et al. Examining the effect of chronic intranasal oxytocin administration on the


neuroanatomy and behavior of three autism-related mouse models. _Neuroimage_ 257, 119243 (2022). Article  CAS  PubMed  Google Scholar  * Wang, Y. et al. Repeated oxytocin prevents central


sensitization by regulating synaptic plasticity via oxytocin receptor in a chronic migraine mouse model. _J. Headache Pain._ 22, 84 (2021). Article  CAS  PubMed  Google Scholar  * Bales, K.


L. et al. Long-term exposure to intranasal oxytocin in a mouse autism model. _Transl. Psychiatry_ 4, e480 (2014). Article  CAS  PubMed  Google Scholar  * Huang, H. et al. Chronic and acute


intranasal oxytocin produce divergent social effects in mice. _Neuropsychopharmacology_ 39, 1102–1114 (2014). Article  CAS  PubMed  Google Scholar  * Zocher, S., Overall, R. W., Lesche, M.,


Dahl, A. & Kempermann, G. Environmental enrichment preserves a young DNA methylation landscape in the aged mouse hippocampus. _Nat. Commun._ 12, 3892 (2021). Article  ADS  CAS  PubMed 


Google Scholar  * Alberca, C. D., Papale, L. A., Madrid, A. & Alisch, R. S. Hippocampal and peripheral blood DNA methylation signatures correlate at the gene and pathway level in a mouse


model of autism. _Hum. Mol. Genet._ 32, 3312–3322 (2023). Article  CAS  PubMed  Google Scholar  * Hadad, N. et al. Absence of genomic hypomethylation or regulation of cytosine-modifying


enzymes with aging in male and female mice. _Epigenetics Chromatin_ 9, 30 (2016). Article  PubMed  Google Scholar  * Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie


2. _Nat. Methods_ 9, 357–359 (2012). Article  CAS  PubMed  Google Scholar  * Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq


applications. _Bioinformatics_ 27, 1571–1572 (2011). Article  CAS  PubMed  Google Scholar  * Danecek, P. et al. Twelve years of SAMtools and BCFtools. _Gigascience_ 10,


https://doi.org/10.1093/gigascience/giab008 (2021). * Akalin, A. et al. methylKit: a comprehensive R package for the analysis of genome-wide DNA methylation profiles. _Genome Biol._ 13, R87


(2012). Article  PubMed  PubMed Central  Google Scholar  * Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. _Bioinformatics_ 26,


841–842 (2010). Article  CAS  PubMed  Google Scholar  * Titus, A. J., Gallimore, R. M., Salas, L. A. & Christensen, B. C. Cell-type deconvolution from DNA methylation: a review of recent


applications. _Hum. Mol. Genet._ 26, R216–R224 (2017). Article  CAS  PubMed  PubMed Central  Google Scholar  * Lee, D., Lee, S. & Kim, S. PRISM: methylation pattern-based,


reference-free inference of subclonal makeup. _Bioinformatics_ 35, i520–i529 (2019). Article  CAS  PubMed  PubMed Central  Google Scholar  * Barrett, J. E. et al. Quantification of tumour


evolution and heterogeneity via Bayesian epiallele detection. _BMC Bioinf._ 18, 354 (2017). Article  Google Scholar  * Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment


analysis web server 2016 update. _Nucleic Acids Res._ 44, W90–97, (2016). Article  CAS  PubMed  PubMed Central  Google Scholar  * Szklarczyk, D. et al. The STRING database in 2021:


customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. _Nucleic Acids Res_ 49, D605–D612 (2021). Article  CAS  PubMed  Google Scholar


  * Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. _Genome Res._ 13, 2498–2504 (2003). Article  CAS  PubMed  PubMed Central


  Google Scholar  * Tang, Y., Zuniga-Hertz, J. P., Han, C., Yu, B. & Cai, D. Multifaceted secretion of htNSC-derived hypothalamic islets induces survival and antidiabetic effect via


peripheral implantation in mice. _Elife_ 9, https://doi.org/10.7554/eLife.52580 (2020). * Zhang, Y., Reichel, J. M., Han, C., Zuniga-Hertz, J. P. & Cai, D. Astrocytic process plasticity


and IKKbeta/NF-kappaB in central control of blood glucose, blood pressure, and body weight. _Cell Metab._ 25, 1091–1102.e1094 (2017). Article  CAS  PubMed  PubMed Central  Google Scholar  *


Ren, J. et al. Intrauterine hyperglycemia exposure results in intergenerational inheritance via DNA methylation reprogramming on F1 PGCs. _Epigenetics Chromatin_ 11, 20 (2018). Article 


PubMed  PubMed Central  Google Scholar  Download references ACKNOWLEDGEMENTS We gratefully thank Cai lab members for technical support, thank W. Freeman and Oklahoma Nathan Shock Center on


Aging for providing kind assistance and Core support, and thank CD Genomics for target methylation sequencing. This research was supported through Einstein institutional resources and partly


through Milky Way Research Foundation award and National Institutes of Health R01AG031774 (all supports to D. Cai). AUTHOR INFORMATION Author notes * These authors contributed equally:


Salman Sadullah Usmani, Hyun-Gug Jung. AUTHORS AND AFFILIATIONS * Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA Salman Sadullah Usmani, Hyun-Gug


Jung, Qichao Zhang, Min Woo Kim, Yuna Choi, Ahmet Burak Caglayan & Dongsheng Cai Authors * Salman Sadullah Usmani View author publications You can also search for this author inPubMed 


Google Scholar * Hyun-Gug Jung View author publications You can also search for this author inPubMed Google Scholar * Qichao Zhang View author publications You can also search for this


author inPubMed Google Scholar * Min Woo Kim View author publications You can also search for this author inPubMed Google Scholar * Yuna Choi View author publications You can also search for


this author inPubMed Google Scholar * Ahmet Burak Caglayan View author publications You can also search for this author inPubMed Google Scholar * Dongsheng Cai View author publications You


can also search for this author inPubMed Google Scholar CONTRIBUTIONS S.S.U. performed all computational analysis on BOCS metadata and target promoter sequencing data, did statistics for all


sequencing data, contributed to data interpretation and provided writing assistance. H.J. performed animal treatment and behavioral study, performed qRT-PCR assays, and contributed to data


interpretation. Q.Z. performed histological and immunostaining experiment and contributed to data interpretation. M.W.K. co-performed animal treatment and behavioral assays. Y.C. contributed


to qRT-PCR primers and target promoter bisulfite sequencing. A.B.C. contributed to animal treatment study. D.C. conceived the hypothesis and ideas, conceptualized and constructed the


project, designed computational and experimental strategies, advised and supervised all experiments, instructed and concluded data analysis, led and concluded data interpretation, and wrote


the paper. All authors approved the paper. CORRESPONDING AUTHOR Correspondence to Dongsheng Cai. ETHICS DECLARATIONS COMPETING INTERESTS D.C. has an invention disclosure with Albert Einstein


College of Medicine (OXT-GnRH approaches for addressing AD or aging). Other authors declare no competing interests. PEER REVIEW PEER REVIEW INFORMATION _Nature Communications_ thanks Isabel


Castanho and the other anonymous reviewer(s) for their contribution to the peer review of this work. A peer review file is available. ADDITIONAL INFORMATION PUBLISHER’S NOTE Springer Nature


remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. SUPPLEMENTARY INFORMATION SUPPLEMENTARY INFORMATION DESCRIPTION OF ADDITIONAL


SUPPLEMENTARY FILES SUPPLEMENTARY DATASET 1 SUPPLEMENTARY DATASET 2 SUPPLEMENTARY DATASET 3 REPORTING SUMMARY SOURCE DATA SOURCE DATA TRANSPARENT PEER REVIEW FILE RIGHTS AND PERMISSIONS OPEN


ACCESS This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and


reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you


modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in


this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative


Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a


copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/. Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Usmani, S.S., Jung, HG., Zhang, Q. _et al._


Targeting the hypothalamus for modeling age-related DNA methylation and developing OXT-GnRH combinational therapy against Alzheimer’s disease-like pathologies in male mouse model. _Nat


Commun_ 15, 9419 (2024). https://doi.org/10.1038/s41467-024-53507-8 Download citation * Received: 28 July 2023 * Accepted: 09 October 2024 * Published: 31 October 2024 * DOI:


https://doi.org/10.1038/s41467-024-53507-8 SHARE THIS ARTICLE Anyone you share the following link with will be able to read this content: Get shareable link Sorry, a shareable link is not


currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative