DNA methylation in the mouse cochlea promotes maturation of supporting cells and contributes to the failure of hair cell regeneration
Edited by Brigid Hogan, Duke University, Durham, NC; received January 20, 2023; accepted July 11, 2023
Significance
Age-related hearing loss can significantly impact quality of life. One potential approach to restore hearing is to regenerate mechanosensory hair cells responsible for detecting sound by the conversion of neighboring supporting cells into new hair cells. However, mammalian supporting cells can only transdifferentiate during embryonic and early postnatal development, and this ability is lost before the onset of hearing. We show that supporting cells accumulate DNA methylation, a form of epigenetic silencing, to permanently shut off the hair cell gene program required for successful transdifferentiation. Blocking ten-eleven translocation (TET) enzyme activity extends the window in which transdifferentiation can occur. Moreover, the loss of hair cells by deafening partially reverses DNA methylation in supporting cells, suggesting one avenue for therapeutic intervention.
Abstract
Mammalian hair cells do not functionally regenerate in adulthood but can regenerate at embryonic and neonatal stages in mice by direct transdifferentiation of neighboring supporting cells into new hair cells. Previous work showed loss of transdifferentiation potential of supporting cells is in part due to H3K4me1 enhancer decommissioning of the hair cell gene regulatory network during the first postnatal week. However, inhibiting this decommissioning only partially preserves transdifferentiation potential. Therefore, we explored other repressive epigenetic modifications that may be responsible for this loss of plasticity. We find supporting cells progressively accumulate DNA methylation at promoters of developmentally regulated hair cell genes. Specifically, DNA methylation overlaps with binding sites of Atoh1, a key transcription factor for hair cell fate. We further show that DNA hypermethylation replaces H3K27me3-mediated repression of hair cell genes in mature supporting cells, and is accompanied by progressive loss of chromatin accessibility, suggestive of facultative heterochromatin formation. Another subset of hair cell loci is hypermethylated in supporting cells, but not in hair cells. Ten-eleven translocation (TET) enzyme-mediated demethylation of these hypermethylated sites is necessary for neonatal supporting cells to transdifferentiate into hair cells. We also observe changes in chromatin accessibility of supporting cell subtypes at the single-cell level with increasing age: Gene programs promoting sensory epithelium development loses chromatin accessibility, in favor of gene programs that promote physiological maturation and function of the cochlea. We also find chromatin accessibility is partially recovered in a chronically deafened mouse model, which holds promise for future translational efforts in hearing restoration.
Sign up for PNAS alerts.
Get alerts for new articles, or get an alert when an article is cited.
The most common cause of hearing loss is the damage and death of mechanosensory cochlear hair cells, which do not functionally regenerate in adult mammals (1, 2). However, nonmammalian vertebrates such as zebrafish and birds can regenerate hair cells throughout life, which occurs through transdifferentiation or conversion of the surrounding supporting cells to hair cells, with or without cell division (3, 4). In contrast, mammals such as mice have only a transient potential for transdifferentiation (5, 6). Beyond this stage, which in mice is just a few days after birth, the mammalian cochlea cannot regenerate hair cells after they have been killed (7, 8).
Transdifferentiation of supporting cells into hair cells is an attractive strategy for hair cell regeneration because both supporting cells and hair cells arise from a common post-mitotic progenitor population during embryonic development (9). Hair cell differentiation begins at E14.5 in mice with the onset of ATOH1 expression, a bHLH transcription factor that is necessary to promote hair cell fate and survival (10, 11). Initially, ATOH1 is broadly expressed in the postmitotic progenitors of the cochlear epithelium (11, 12). Subsequent upregulation of the Notch ligands DLL1 and JAG2 by differentiating hair cells triggers Notch-mediated lateral inhibition of Atoh1 expression in adjacent cells and directs them toward a supporting cell fate (13, 14).
During the first postnatal week, mouse cochlear supporting cells retain some plasticity to transdifferentiate into hair cells, suggesting they may be maintained in a progenitor-like state (15, 16). This change from a cell state that maintains regenerative ability to the mature cell state in the functional, hearing cochlea is driven by both transcriptional and epigenetic changes. We previously showed that one mechanism that partly explains the loss of transdifferentiation potential is the decommissioning of hair cell enhancers in maturing supporting cells marked by H3K4me1 removal (17). However, other repressive epigenetic mechanisms such as heterochromatin formation are known to further silence genes of alternative cell identities, and reinforce cell fate commitment during development and maturation (18, 19).
We hypothesized that hair cell genes are further silenced in supporting cells by heterochromatin formation during their maturation. One of the hallmarks of heterochromatin is DNA methylation at CpG dinucleotides (20–22), which has been shown to restrict pluripotency in progenitor cells by preventing transcription factor binding and permanently silencing genes (23, 24). In the present study, we first characterized changes in DNA methylation of supporting cells between birth and weaning at postnatal day 21. We found that de novo CpG methylation encroaches on DNA methylation “valleys” of developmentally regulated hair cell gene promoters in supporting cells, and that this increase in DNA methylation coincides with the loss of chromatin accessibility in supporting cells. We also found that supporting cells undergoing transdifferentiation induce de novo DNA demethylation on a set of hair cell–specific regulatory elements, which led us to postulate that the active removal of DNA methylation by ten-eleven translocation (TET) enzyme activity is required for the transdifferentiation of a postmitotic supporting cell. We also found that hair cell genes with CpG island–associated promoters continue to lose chromatin accessibility at the single-cell level in every supporting cell subtype as they reach adulthood. Our work provides an epigenetic analysis of heterochromatin formation in the inner ear as the cochlea matures and loses its regenerative ability.
Results
Cochlear Progenitor Cells, Hair Cells, and Supporting Cells Have Distinct DNA Methylation Signatures.
We hypothesized that systemic heterochromatinization contributes to the loss of transdifferentiation potential in supporting cells. To identify genomic regions undergoing heterochromatinization in maturing postnatal supporting cells, we examined DNA methylation, a feature of heterochromatin. We profiled DNA methylation using whole-genome bisulfite sequencing (WGBS) on cells from the organ of Corti at the following key developmental stages: embryonic day 13.5 prosensory progenitor cells (E13.5 PG), postnatal day 1 hair cells (P1 HC), postnatal day 1 supporting cells (P1 SC), postnatal day 6 supporting cells (P6 SC), and postnatal day 21 supporting cells (P21 SC). We used enzymatic digestion followed by fluorescence-activated cell sorting (FACS) mediated-purification to isolate E13.5 prosensory progenitors from p27Kip1-GFP mice, P1 hair cells from Atoh1-GFP mice, and P1 supporting cells from Lfng-EGFP mice. p27Kip1-GFP was used to isolate prosensory progenitor cells at E13.5 that will eventually give rise to both hair cells and supporting cells during development (9, 25). For P6 and P21 supporting cells, we used the NuTRAP homogenization method to isolate nuclei from Lfng-CreER; NuTRAP mice (SI Appendix, Fig. S1 and Materials and Methods). We refer to promoters of genes that sit within a CpG island as CGI promoters, and promoters of genes that do not sit within a CpG island as non-CGI promoters. CGIs are genomic regions of 500 to 1,500 bp long with a ratio of CpG dinucleotide sequences greater than 0.6, and they are often found at promoters and retrotransposable elements (26). As a positive control, we confirmed that DNA methylation is depleted at 13,564 CGI promoters and enriched at 8,586 non-CGI promoters (SI Appendix, Fig. S2 A and D). We observed distinct methylomes in the different cell types and time points. For example, we observed that the Atoh1 and Dll1 loci are more heavily methylated in P21 supporting cells compared to E13.5 prosensory progenitors, P1 hair cells, P1 supporting cells, and P6 supporting cells (Fig. 1A). Since the ATOH1 transcription factor is both necessary and sufficient for hair cell differentiation, we profiled ATOH1 chromatin-binding regions in E17.5 hair cells. We used CUT&RUN with anti-GFP antibodies to identify binding sites in hair cells obtained from Atoh1-EGFP mice. We found previously identified ATOH1-binding sites at the Atoh1 promoter and its 3′ autoregulatory enhancer overlapping with regions of DNA hypermethylation in P21 supporting cells (Fig. 1A). We saw a similar pattern at the Dll1 locus, which is specifically expressed in hair cells (Fig. 1A). DNA hypermethylation at these regulatory elements could serve as a mechanism to permanently silence the endogenous Atoh1 and Dll1 loci in mature supporting cells by preventing ATOH1 binding. Since ATOH1 and DLL1 play critical roles in hair cell and supporting cell fate specification, DNA hypermethylation of these gene loci suggests a larger genome-wide shutdown of gene programs responsible for the development of hair cells and supporting cells with increasing age.
Fig. 1.

We characterized the changes in DNA methylation on a genome-wide level in our samples using principal component analysis (PCA). We separated our DNA methylation data into different genomic features—CpG islands, promoters, and putative enhancers—prior to PCA analysis (Fig. 1B). Annotations of CpG islands and promoters were obtained from the UCSC Genome Annotation Database for GRCm38/mm10 (27). Putative enhancers were identified by calling peaks on H3K4me1 CUT&Tag signal (Fig. 1D). At CpG islands, E13.5 prosensory progenitors, P1 hair cells, and P1 supporting cells clustered together, while P6 supporting cells and P21 supporting cells clustered away from all other cell types and stages, and also from each other (Fig. 1 B, Left). This clustering pattern suggests that DNA methylation at CpG islands stays relatively constant among cochlear cell types at earlier stages of development, but diverges at the end of the first postnatal week. At gene promoters, all cell types and stages clustered separately, with E13.5 prosensory progenitors in the middle and hair cells and supporting cells diverging toward separate identities (Fig. 1 B, Middle). This suggests that there are distinguishing DNA methylation patterns at promoters between specific cell types and stages of development in the organ of Corti. At gene enhancers, P1 hair cells and P1 supporting cells cluster away from the prosensory progenitor state but are relatively similar to each other, suggesting they still share similar enhancer networks at this earlier stage of development, consistent with our previous data (Fig. 1 B, Right) (17). Overall, our data suggest that DNA methylation is positively correlated with the increasing maturation of supporting cells with age.
Next, we sought to identify the genomic regions driving the variance between the cell types that could contribute to distinct gene silencing patterns. We used dmrseq to identify differentially methylated regions (DMRs). We identified DMRs between E13.5 prosensory progenitors and P1 hair cells, P1 hair cells and P1 supporting cells, and P1 supporting cells and P21 supporting cells to obtain sets of regions that were hypermethylated or hypomethylated in one group versus the other. We further intersected the DMR sets to obtain four mutually exclusive sets of regions, which we named according to the dynamics of their DNA methylation signature (Fig. 1C). All four sets of DMRs showed evolutionary conservation, suggesting that they possess some regulatory function and giving confidence to the peak calling method (SI Appendix, Fig. S2C). We identified 2,850 preestablished DMRs, which are characterized by being hypomethylated in E13.5 prosensory progenitors, P1 hair cells and P1 supporting cells, becoming progressively hypermethylated in P6 and P21 supporting cells. We identified 6,746 de novo hair cell–specific DMRs, which are uniquely hypomethylated in P1 hair cells. We identified 3,058 de novo supporting cell–specific DMRs, which become most heavily hypomethylated in P21 supporting cells. Finally, we identified 9,683 de novo common DMRs, which are hypomethylated in P1 hair cells, P1 supporting cells, P6 supporting cells, and P21 supporting cells (Fig. 1C). Both the preestablished and de novo hair cell–specific DMRs showed enrichment of the ATOH1 E-Box and POU4F3 homeobox motifs, suggesting that these DMRs are part of the hair cell gene regulatory network (SI Appendix, Fig. S2B). Strikingly, the DMRs appear to be highly specific to organ of Corti cells, as the DMRs are completely hypermethylated in intestinal tissues, even though both tissues express ATOH1 and regulate cell fate by Notch signaling (SI Appendix, Fig. S2E) (28–30). The de novo distinction we used is important, because the prosensory progenitors, hair cells, and supporting cells are postmitotic at all developmental stages examined in this study. Thus, the mechanism of DNA hypomethylation is most likely actively mediated by TET enzyme activity, rather than by passive DNA demethylation during cell division (31, 32). Taken together, our analysis shows that DNA methylation patterns coincide with distinct developmental stages and cell types.
DNA Methylation Signatures Highlight Gene Regulatory Network Switching within the Prosensory Epithelium as it Matures.
To determine if the DMRs identified in our analysis were part of the hair cell gene regulatory network, we overlaid the DMR sets with binding sites for two key hair cell transcription factors, ATOH1 and POU4F3. We used published ATOH1 and POU4F3 CUT&RUN data from FACS-purified E17 hair cells (17, 33) (Fig. 1D). POU4F3 is a downstream target of ATOH1 and has pioneer transcription factor activity required for accessing closed ATOH1 target enhancers and allowing hair cell differentiation to proceed (33). The de novo hair cell–specific DMRs and the de novo common DMRs contain both ATOH1- and POU4F3-binding sites, suggesting that they might be downstream targets of ATOH1 and POU4F3, and are therefore required for the differentiation of hair cells. On the other hand, the preestablished DMRs and the de novo supporting cell–specific DMRs are only bound by ATOH1, and not by POU4F3 (Fig. 1D). Since the preestablished DMRs are hypomethylated in E13.5 prosensory progenitors, we suggest these regions may serve as initial targets of ATOH1, which are up-regulated within the prosensory domain at E14.5 to initiate hair cell differentiation (10, 33). To further categorize these DMR sets, we performed CUT&TAG on the promoter-specific mark H3K4me3 and the promoter-enhancer mark H3K4me1 in P1 hair cells and P1 supporting cells (34). We found the DMR sets are depleted of the promoter mark H3K4me3 but are enriched for the enhancer mark H3K4me1 in both hair cells and supporting cells (Fig. 1D), suggesting that the DMRs are putative hair cell–specific and supporting cell–specific enhancers.
We next performed gene ontology (GO) enrichment analysis on the DMR sets using GREAT (35). The preestablished DMRs include known organ of Corti developmental genes such as Sox2, Atoh1, Gfi1, Notch1, and Dll1. The preestablished DMRs are enriched for GO terms relating to “regulation of epithelial cell differentiation,” “stem cell population maintenance,” and “regulation of auditory receptor cell differentiation” (Fig. 1E). The de novo hair cell–specific DMRs are enriched for terms relating to the maturation of hair cells, such as “neuromuscular process controlling balance,” “mitochondrial membrane organization”, and “auditory receptor cell stereocilium organization”. Genes within the de novo hair cell–specific DMRs include early hair cell developmental genes like Atoh1, Pou4f3, Gfi1, Myo7a, Myo6, and Jag2, as well as genes involved in the maturation of hair cells such as Pcdh15, Cdh23, and Clic5. The de novo supporting cell–specific DMRs have GO terms relating to epithelium formation with terms such as “cell–cell junction organization,” and “positive regulation of cytoskeleton organization.” Key genes include Sox2, Sox4, Sox9, Gjb2, Gjb6, Coch, KCN (potassium channel) family genes, and SLC (solute carrier) family genes. Finally, the de novo common DMRs have GO terms relating to more general epithelial maturation, such as “cytoskeleton-dependent intracellular transport” and “adherens junction organization” and broadly include KCN, SLC, and CDH (cadherin) family genes. The GO analyses demonstrate that each organ of Corti cell type has a distinct DNA methylation signature at each age that correlates with its known gene expression and function. Taken together, we suggest that dynamic DNA methylation patterns are used to reinforce gene program switches as cells transition through different states and identities during their lifetime. For instance, as supporting cells lose the ability to transdifferentiate postnatally, we observed a switch from a hair cell differentiation gene program to a supporting cell maturation gene program, where the former becomes hypermethylated and the latter becomes hypomethylated.
DNA Methylation Encroachment and Heterochromatinization of Preestablished CpG Island Promoters Occurs in Postnatally Maturing Supporting Cells.
The gradual DNA hypermethylation of preestablished DMRs in postnatal supporting cells during the first postnatal week correlates with the period during which supporting cells lose the potential to transdifferentiate in response to DAPT (N-[N-(3, 5-difluorophenacetyl)-l-alanyl]-s-phenylglycinet-butyl ester) (13, 36). Initially, the percentage of methylated CpGs at preestablished DMRs decreased from 45% in E13.5 prosensory progenitors to 30% in P1 hair cells and P1 supporting cells, which coincided with the start of differentiation of cochlear hair cells and supporting cells (Fig. 2A). The 30% mCpG, although an average of all preestablished DMRs, corresponds to the lowest point of the DNA methylation valleys observed at the Atoh1, Pou4f3, and Gfi1 loci (Fig. 2D). The CpG percent methylation then increased gradually to 38% and 50% by P6 and P21 in supporting cells, coinciding with loss of transdifferentiation potential (Fig. 2A). To better understand the role of DNA hypermethylation in supporting cells, we annotated the preestablished DMRs to genomic features (37), and found that they are enriched around CpG islands and their corresponding shores and shelves, which are defined as iterative 2 kb regions flanking the CpG island (Fig. 2 B and C). In agreement with this, the preestablished DMRs are also enriched around genic regions, spanning from 5 kb upstream of the promoter, through the gene body, and all the way to the 3′ UTR (Fig. 2C). The preestablished DMRs are not enriched at intergenic regions, suggesting that they are unlikely retrotransposons or distal enhancers. Rather, the majority of preestablished DMRs appear to identify proximal and genic enhancers that occur within the neighborhood of CpG island–containing promoters, but not necessarily occurring at the promoter itself since they do not possess H3K4me3 peaks. Therefore, we focused our attention on preestablished DMRs overlapping CGI promoters in our subsequent analyses.
Fig. 2.

CGI promoter genes serve important roles as transcriptional hubs and tend to be developmentally regulated. They sit within DNA methylation valleys, also referred to as DNA methylation canyons, which are largely hypomethylated (38). These CGI promoter genes are frequently silenced by the repressive histone modification H3K27me3. The recruitment of PRC2, a H3K27me3-specific methyltransferase, to CG rich sequences at promoters also repels DNA methyltransferases (DNMTs) (39). Thus, CGI promoters, used for lineage specification during the process of cell differentiation, are maintained in a DNA hypomethylated state in differentiated cells. CGI promoter repression is understood to be mediated by H3K27me3 for both the prelineage commitment bivalent state, as well as the subsequent repression of nonlineage genes (40). DNA hypermethylation has been shown to occur at promoters that are CG-poor and lack CGIs, which tend to be somatic tissue–specific genes expressed in more mature cell types (38). Building on this model, we found that cochlear supporting cells undergo a switch between the two modes of repression—from H3K27me3 methylation to DNA methylation—at developmentally regulated hair cell–specific CGI promoter genes (Fig. 2D). For example, at three key hair cell–specific transcription factor gene loci, Atoh1, Pou4f3, and Gfi1, we find evidence of both DNA hypermethylation and heterochromatinization. Additional examples of DNA hypermethylation were observed at Tead4, Eya1, Notch1, and Fgfr3 (SI Appendix, Fig. S3A). At the genic level, the DNA hypermethylation of preestablished DMRs manifests as DNA methylation encroachment of the DNA methylation valleys in which the respective CGI promoter gene sits (Fig. 2D, WBGS).
Next, we identified the epigenetic changes occurring across 103 gene promoters that overlapped with the previously identified set of 2,850 preestablished DMRs (Fig. 2 E–G). We observed a switch from H3K27me3-mediated repression to a DNA methylation–mediated silencing in supporting cells between P6-P8 and P21 (Figs. 2E and 3F). This finding agrees with previous work describing the antagonistic relationship between DNA methylation and PRC2-mediated H3K27me3 deposition (41, 42). The switch from H3K27me3-mediated repression to DNA methylation–mediated silencing was also observed at both HoxA and HoxB loci (SI Appendix, Fig. S4B). In addition to DNA hypermethylation, DNA accessibility measured by Cleavage Under Targeted Accessible Chromatin (CUTAC) (43) drops precipitously between P1 and P21 in supporting cells (Fig. 2G). While the increase of H3K27me3 between P1 and P8 explains the decreased accessibility within that same time window, DNA hypermethylation explains the decrease in accessibility between P8 and P21, suggesting that DNA hypermethylation is required in the final stages of maturation of supporting cells to complete heterochromatinization of their hair cell–specific CGI promoters. Besides CGI promoters, we also see similar changes in DNA methylation, H3K27me3, and accessibility at pre-established DMRs that overlap with non-CGI promoters, gene bodies, and intergenic regions, albeit less prominently (SI Appendix, Fig. S3B). As a positive control, we confirmed that H3K27me3 signal at promoters is comparable across samples (SI Appendix, Fig. S4A). Altogether, we propose switching between H3K27me3 and DNA methylation is a mechanism for long-term silencing of hair cell–specific CGI promoter genes and the complete shutting down of the hair cell gene regulatory network.
Fig. 3.

DAPT-Mediated Transdifferentiation Induces De Novo DNA Demethylation of Hair Cell DMRs.
Neonatal supporting cells still retain the potential to transdifferentiate into hair cells after hair cell loss, or by blocking Notch signaling with the gamma-secretase inhibitor DAPT (8, 13, 36, 44, 45). However, from our DNA methylation data, we observe that the de novo hair cell–specific DMRs are hypermethylated in P1 supporting cells (Fig. 1C). Based on the previously described GO terms, the hair cell–specific DMRs are essential for hair cell differentiation (Fig. 1E). Since DNA methylation is a repressive epigenetic feature, we hypothesized that in cases where P1 supporting cells differentiate into hair cells, hypermethylated hair cell–specific DMRs in P1 supporting cells must first undergo demethylation and derepression to successfully up-regulate hair cell–specific genes and transdifferentiate. To test this, we induced P0 supporting cell transdifferentiation with DAPT and performed WGBS to measure mCpG percentages in DAPT-responsive supporting cells compared to DAPT-unresponsive cells. Briefly, we used a transgenic mouse line with Lfng-CreERT2 (46), and a ROSA tdTomato (TDT) Cre reporter (47) to lineage trace supporting cells, as well as Atoh1-GFP (48) to label hair cells and supporting cells undergoing transdifferentiation into new hair cells (17). Lineage-traced TDT+ supporting cells that transdifferentiate into hair cells upon DAPT treatment up-regulate hair cell–specific transcription factors ATOH1-GFP and POU4F3 (Fig. 3A and SI Appendix, Fig. S5A). Thus, supporting cells undergoing transdifferentiation exhibit TDT+/ GFP+ double labeling, whereas supporting cells not responding to DAPT are only labeled with the TDT reporter (Fig. 3A). Cochlear sensory epithelium was explanted and cultured with or without DAPT for 48 h, followed by FACS purification of single-labeled nonresponsive TDT+ supporting cells and double-labeled TDT+/GFP+ transdifferentiated supporting cells (Fig. 3 B and C). We processed these purified cells for bisulfite treatment for WGBS analysis. We also performed this experiment in supporting cells at P6 when the supporting cells no longer respond to DAPT.
To establish a baseline, we compared the CpG methylation percentage at de novo hair cell–specific DMRs to endogenous P1 supporting cells (Fig. 3D). After DAPT treatment, we found that only successfully transdifferentiated supporting cells at P0 showed a decrease in mCpG percentage at de novo hair cell–specific DMRs (Fig. 3D). No decrease in mCpG percentage was observed at the preestablished, de novo supporting cell–specific, or de novo common DMRs, suggesting that the demethylation is highly specific (SI Appendix, Fig. S5B). Nonresponsive TDT+ supporting cells in both the P0 and P6 cochlear explants had comparable percentage of mCpG to baseline in vivo P1 supporting cells. Quantifying the change, we found a 10% decrease in mCpG percentage from 72.37% in nonresponsive TDT+ supporting cells to 62.33% in transdifferentiated TDT+/GFP+ supporting cells (P-value: 2.97e-61; Fig. 3E). The 10% decrease in DNA methylation is notable because the demethylation process occurred within a relatively brief window of 48 h after DAPT exposure. In addition, earlier studies demonstrate that 5 to 10% DNA methylation differences can be biologically significant, for example in the case of the Oct4 locus between human embryonic stem cells, their derived cell populations, normal somatic tissues, and disease conditions (20, 49). We used de novo common regions as a control and found no significant change in DNA methylation levels (P-value: 1; Fig. 3E). The preestablished DMRs and the de novo supporting cell–specific DMRs also show no signs of demethylation (SI Appendix, Fig. S5B), indicating that the DNA demethylation is specific to the de novo hair cell–specific DMRs. Since the transdifferentiating TDT+/GFP+ supporting cells are postmitotic, the only mode for de novo DNA demethylation is through TET enzyme activity, and not through the lack of maintenance by DNMT1 during cell division (50). Together, this suggests that the activation of the hair cell GRN in transdifferentiating supporting cells is partly mediated by DNA demethylation of hair cell–specific DMRs, which is most likely driven by TET enzyme activity.
TET enzymes are responsible for the stepwise conversion of 5-methylcytosine to 5-hydroxymethylcytosine to 5-formylcytosine to 5-carboxylcytosine (51). The action of thymine-DNA-glycosylase (TDG) and base excision repair (BER) ultimately restores the unmethylated cytosine (51). The WGBS method we use detects both 5-methylcytosine and 5-hydroxymethylcytosine. Since we detected DNA demethylation after supporting cell transdifferentiation, we concluded that the 5-methylcytosine is being converted to at least 5-formylcytosine by TET enzyme activity, which may be sufficient to derepress hair cell–specific regulatory elements. We tested whether TET-mediated DNA demethylation is required for supporting cells to transdifferentiate into hair cells. We treated P0 cochlear explants with 10 µM of DAPT and a dose series of Bobcat339 (52), a TET1 and TET2 enzyme inhibitor for 48 h. Cochleas were then dissociated after 48 h for FACS-mediated quantification. We hypothesized that blocking TET enzyme activity with Bobcat339 would prevent supporting cells from successfully up-regulating ATOH1 and transdifferentiating in response to DAPT. This would lead to a decrease in the proportion of double-labeled GFP+/TDT+ cells (which we quantified as % permissive supporting cells) compared to total TDT+ cells. In response to 10 µM DAPT treatment, 56.97% of supporting cells were permissive to transdifferentiation. However, supporting cell transdifferentiation rate decreased in a dose-dependent manner with increasing concentrations of Bobcat339 cotreatment (Fig. 3F). We found 45.00% permissive supporting cells with 90 µM Bobcat339 cotreatment (n = 3, P-value = 0.085, n.s.), 30.09% permissive supporting cells with 180 µM Bobcat339 cotreatment (n = 3, P-value = 0.023, *), and 14.94% permissive supporting cells with 270 µM Bobcat339 cotreatment (n = 3, P-value = 0.030). This demonstrates that supporting cells require TET enzyme activity to demethylate and derepress hair cell–specific DMRs before they can be activated during transdifferentiation.
Single-Cell Profiling Demonstrates a Developmental Switch in Supporting Cell Gene Networks, and Changes in Heterochromatin Landscape in the Deafened Adult Cochlea.
DNA methylation is considered to be the final layer of epigenetic silencing to permanently shut down DNA and make it refractory to transcription factors and transcriptional machinery (53). We have demonstrated that preestablished CpG island promoters undergo increasing de novo DNA methylation and decreasing accessibility in postnatal maturing supporting cells (Fig. 2D). Further, we showed that the transdifferentiation of P1 supporting cells both induces de novo DNA demethylation (Fig. 3D) and is attenuated by inhibiting TET enzyme activity (Fig. 3F), suggesting that DNA methylation plays a key role in silencing the hair cell gene regulatory network through heterochromatin formation. Next, we wanted to understand the implications of this heterochromatinization in the pathological context of hearing loss. Specifically, we wanted to understand whether there are further changes to the supporting cell chromatin landscape in the adult stage, as well as in a long-term deafened state, which is clinically relevant to human hearing loss. Since supporting cell subtypes are functionally distinct at adult stages, we used scMultiome to simultaneously profile gene expression and chromatin accessibility at the single-cell level. Comparing accessibility between organ of Corti supporting cell subtypes allows us to identify cell type–specific enhancers, as well as supporting cells that are more receptive to conversion into new hair cells. We collected scMultiome datasets for cochlear supporting cells in wildtype P1, P8 (54), and P70 mice, as well as in P70 mice that were deafened at P21 (Fig. 4A). For the P70 dataset, Lfng-CreERT2;NuTRAP;Pou4f3-DTR mice were used to label and sort supporting cells. Single-cell data were clustered using “uniform manifold approximation and projection” (UMAP) and “weighted-nearest neighbor” (WNN) using the Seurat R package, which integrates both the gene expression and the accessibility information to define a “joint” cellular state (55). WNN analysis is an improvement over previous methods of clustering that were based on RNA or ATAC data alone by improving our ability to resolve cell states (SI Appendix, Fig. S6A). scMultiomic profiling is especially well-suited for resolving the diversity of postnatal organ of Corti supporting cells, which no longer express distinguishing developmental genes and instead share common morphological and molecular features (56).
Fig. 4.

We annotated the clusters using gene expression data cross-referenced with previously published literature. Since gene programs change as the cochlea matures between P1, P8, and P70, we used different sets of genes to identify clusters at each developmental time point. Genes used to identify clusters from the P1, P8, and P70 cochlea 10× multiome datasets can be found in SI Appendix, Figs. S7–S9. After using the single-cell gene expression data to identify and annotate clusters, we compared the single-cell ATAC data between clusters and time points. We found that cochlear developmental genes such as Atoh1 were more accessible at P1 in both hair cells and supporting cells, whereas genes important for the physiological function of the cochlea such as Gjb2 and Gjb6 were more accessible at P70 in supporting cells (Fig. 4B). Altogether, this suggests that maturing supporting cells undergo a gene program switch, where the hair cell gene program is silenced in favor of a differentiated and functional supporting cell gene program. Additional examples of hair cell–specific genes Pou4f3, Jag2, Dll1, and Myo7a, and supporting cell–specific genes Sox2, Notch1, and Jag1 are shown in SI Appendix, Figs. S9B and S10A, respectively.
Next, we associated the single-cell ATAC data back to our DNA methylation data. We separated chromatin accessibility (ATAC) signal by cell types, and quantified it over the preestablished, hair cell–specific, supporting cell–specific, and common DMRs (Fig. 5A). We found supporting cell subtypes showed progressively decreasing accessibility at preestablished and hair cell–specific DMRs between P1, P8, and P70 time points, but not at the SC-specific or common DMRs (Fig. 5A). We noted that accessibility increased in the P70 long-term deafened supporting cells compared to the P70 wild-type supporting cells at hair cell–specific DMRs in BC, PC, and DC2 clusters (Fig. 5A). The most prominent example of this can be seen at the locus of the hair cell gene Myo7a, where increased accessibility in the deafened BC cluster also correlated with low Myo7a mRNA expression (Fig. 5B). To validate the Myo7a mRNA detected in the deafened BC cluster, we performed RNAscope RNA FISH. We identified Epyc as a putative BC marker from our P70 10× multiome dataset and confirmed the expression of Epyc in BCs in intact tissue using RNAscope (Fig. 5 C and D). We were able to detect Myo7a transcripts coming from the surviving Epyc+ BC after hair cell killing (Fig. 5D). This suggests that the loss of hair cells may cause supporting cells to modify their chromatin structure following damage, which may allow for easier reactivation of the hair cell GRN in the deafened state.
Fig. 5.

Discussion
As cells transition from totipotent and pluripotent states to different cell lineages, gene networks are activated to convert cells to progressively more restricted cell identities. Concurrently, repressive epigenetic barriers are deployed to silence gene networks of alternative cell lineages and cell fates. This maturation process is critical for maintaining the fidelity of tissue function and preventing the uncontrolled growth of cancers. The processes of terminal differentiation and maturation frequently militate against regeneration in mammals, and this age-dependent decline in regenerative potential has been well characterized in tissues such as skin, brain, heart, and muscle. Like cells in many other tissues, cochlear mechanosensory hair cells do not regenerate in adulthood, and demonstrate only a limited regeneration potential in the first postnatal week of mice through direct transdifferentiation of neighboring supporting cells (5, 6). Previous studies have characterized transcriptional and epigenetic changes such as enhancer decommissioning that recapitulate this transition from regenerative ability to maturation and loss of plasticity (17, 58, 59).
It is notable that decommissioned enhancers are not permanently silenced: They can still be primed and reactivated if the CpG dinucleotides within the enhancer region are retained in a hypomethylated state (30, 60, 61). A combination of enhancer decommissioning, acquisition of repressive histone marks, and DNA methylation is therefore more likely to place lineage-specific genes in a more inaccessible state. In the present study, we demonstrated that some enhancers bound by the hair cell transcription factor ATOH1 that reside in DNA methylation “valleys” of hair cell–specific genes are hypermethylated as supporting cells undergo maturation (Fig. 1C). DNA methylation of CpGs is a mechanism to permanently silence regulatory elements by either creating steric hindrance between the transcription factor and its binding motif (24, 62) or by promoting chromatin condensation and heterochromatin formation (53, 63). Moreover, we show evidence of heterochromatin formation, where hair cell enhancers that have acquired H3K27me3 repressive marks further transition to CpG methylation–based silencing, accompanied by complete loss of chromatin accessibility (Fig. 2 E–G). Thus, for a subset of enhancers, hypermethylation of hair cell–specific genes is sufficient for permanent silencing in mature supporting cells. We postulate that this heterochromatin formation impedes ATOH1 binding and prevents, at least in part, the activation of the hair cell gene program in mature supporting cells. Ultimately, DNA methylation silences genes of alternative cell fates and restricts pluripotency in progenitor cells as they differentiate and mature (23, 24). In the case of maturing supporting cells, this can be seen as a transition from a plastic, progenitor-like state to a state where transdifferentiation potential is lost (16).
In addition to supporting cells increasing DNA methylation of hair cell loci with age, we also observed evidence for active regulation of DNA methylation of some hair cell loci in young supporting cells. We found that neonatal supporting cells undergoing transdifferentiation after Notch inhibition showed de novo DNA demethylation at a set of activated hair cell–specific enhancers (Fig. 3D) (17), suggesting that DNA methylation is already present at some hair cell loci in young supporting cells, but that this methylation is reversible. We show that inhibiting TET enzyme activity is sufficient to prevent transdifferentiation of young supporting cells (Fig. 3F). These data show that DNA methylation is already being deployed to silence hair cell enhancers in young supporting cells, but that it is plastic, such that DNA demethylation of these enhancers is required for transdifferentiation into hair cells.
In addition to characterizing changes in DNA methylation as supporting cells mature, we also sought to understand how the chromatin epigenetic landscape continues to change in the adult, well beyond this developmental period. Our scRNA-seq and scATAC-seq multiomics analysis shows that all cochlear supporting cell types continue to lose chromatin accessibility at hair cell–specific enhancers near CpG island promoter genes over the first 10 wk in mice, after both the onset of hearing and sexual maturity, but that mice deafened for 7 wk showed partial recovery of chromatin accessibility at these enhancers (Fig. 5A). Furthermore, the border cell from deafened cochleas with its loosened chromatin partially turns on Myo7a expression (Figs. 5 B and D). This suggests that the border cell may be a potential candidate to drive expression of hair cell–specific genes for regenerative therapies.
However, some qualifications should be considered with this finding. Our model of deafening uses the Pou4f3DTR mouse to specifically kill hair cells after diphtheria toxin treatment; here the mechanism of killing is through termination of protein synthesis (64). This differs from traditional hair cell killing models that use ototoxic drugs such as aminoglycosides or platinum-based chemotherapeutic agents, whose mechanism of action relies on oxidative stress (65). Ototoxic agents may not be entirely specific to hair cells as compared to the Pou4f3DTR approach and may present some toxicity to other cell types such as supporting cells (66). This additional insult to supporting cells may make them even less responsive to regenerative therapies.
Our data suggest that the loss of hair cells elicits a different damage mechanism than the DNA damage response documented in other systems, wherein repressive DNA methylation and histone methylation tend to be recruited to the DNA damage site to temporarily shut down transcription to allow for DNA repair mechanisms to take place (67, 68). In the case of the aging and deafened cochlea, we observe repressive methylation accumulating with age, which coincides with loss of chromatin accessibility. Upon hair cell death and damage to the cochlea, we observe an increase in chromatin accessibility along with Myo7a mRNA transcripts being produced by the border cell population, suggesting that the border cell is becoming more transcriptionally active, rather than being repressed in response to damage.
Our current study has shown that DNA methylation of hair cell loci correlates with the failure of adult supporting cells to regenerate hair cells. However, an additional consideration for hair cell regeneration is whether downregulation of the mature supporting cell gene program is required prior to successful regeneration of hair cells from supporting cells. As cells terminally differentiate, their access to alternative cell type gene programs become limited (69, 70). Forcing the activation of a hair cell gene program–for example, by transcription factor reprogramming (54, 71)—while the supporting cell gene program is still active may prove detrimental. Moreover, although we show some evidence of hair cell gene loci becoming more accessible in the supporting cells of deafened mice, hair cell loci containing binding sites for key transcription factors responsible for establishing hair cell identity such as ATOH1, POU4f3, and GFI1 remain closed in mature supporting cells (Fig. 4B and SI Appendix, Fig. S10). Thus, directed reprogramming using hair cell transcription factors must be accompanied by the manipulation of epigenetic modifications for functional regeneration of hair cells in the deafened cochlea (72–74).
Materials and Methods
A detailed description is included in SI Appendix of this paper. Animals: Experiments were conducted in accordance with the policies of the Institutional Animal Care and Use Committee of the Keck School of Medicine of the University of Southern California. Briefly, cochlear dissections were performed in ice-cold-modified artificial cerebrospinal fluid solution (ACSF) on top of an ice block to provide continuous chilling. For explant culture, the cochlear sensory epithelium was cultured on SPI black filter membranes. To initiate Lfng-CreER-mediated lineage tracing of supporting cells, (Z)-4-Hydroxytamoxifen (Sigma H7904) was added to the culture media. For transdifferentiation experiments, DAPT was added to the culture media. In the TET inhibition experiments, Bobcat339 was added to the culture media. Cochlear sensory epithelia were placed into a 1-mL Dounce tissue grinder with 1 mL of homogenization buffer. The tissue was homogenized with a chilled loose pestle, followed by a chilled tight pestle. The homogenate was filtered through a 40-µm cell strainer and spun down at 1,000 × g and 4 °C for 10 min and resuspended in 1 mL of nuclei wash buffer for FACS purification. Cryosectioning, immunostaining, and microscopy were done as previously described in Tao et al. (17). RNAscope methods are described in SI Appendix, Materials and Methods. To generate P70 long-term deafened mice, we used a Lfng-CreERT2;NuTRAP;Pou4f3DTR mouse to lineage trace SCs and specifically kill hair cells after diphtheria toxin treatment. Lfng-CreERT2;NuTRAP mice were used as wild-type controls. Mice received tamoxifen at P21, Diptheria toxin (Sigma-Aldrich D0564) at P28, and allowed to mature to P70, at which point cochlear tissues were harvested for input into a scMultiome reaction. Next-generation sequencing methods such as WGBS, CUT&RUN, CUT&TAG, scMultiome are described in SI Appendix, Materials and Methods. Sequencing data have been uploaded to the GEO database (https://www.ncbi.nlm.nih.gov/geo), accession number GSE224563.
Data, Materials, and Software Availability
Acknowledgments
We thank Welly Makmura for excellent technical assistance and Francis James for helping set up NGS alignment pipelines and providing auxiliary bioinformatics analysis. We thank past and present Lab members Ksenia Gnedeva, Litao Tao, Robert Rainey, Haoze Yu, Xizi Wang, Louise Menendez, Talon Trecek, Tuo Shi, and Jennifer Rubio for scientific discussions, Veronica Ortiz for providing initial guidance on whole-genome bisulfite sequencing (WGBS) and George Ye and Unmesh Jadhav for providing their published WGBS dataset from intestinal tissue. We are also very grateful for assistance from Jeffrey Boyd and Bernadette Masinsin of the Broad Center Flow Cytometry Facility, Seth Ruffins of the Broad Center Optical Imaging Facility, and David Ruble of the Genomics Core at Children’s Hospital Los Angeles Center for Personalized Medicine. This work was supported by F31 DC018703 to J.D.N., T32 HD060549 (supported J.D.N.)., RO1 DC014832 to A.K.G., RO1 DC015829 to N.S., and a Hearing Restoration Project Consortium award from the Hearing Health Foundation to A.K.G. and N.S.
Author contributions
J.D.N., J.L., J.G.C., and N.S. designed research; J.D.N., J.L., and T.S. performed research; J.D.N. and J.L. contributed new reagents/analytic tools; J.D.N., T.S., A.K.G., and N.S. analyzed data; and J.D.N., A.K.G., and N.S. wrote the paper.
Competing interests
The authors declare no competing interest.
Supporting Information
Appendix 01 (PDF)
- Download
- 5.58 MB
References
1
D. N. Furness, Molecular basis of hair cell loss. Cell Tissue Res. 361, 387–399 (2015).
2
E. L. Wagner, J.-B. Shin, Mechanisms of hair cell damage and repair. Trends Neurosci. 42, 414–424 (2019).
3
J. A. Harris et al., Neomycin-induced hair cell death and rapid regeneration in the lateral line of zebrafish (Danio Rerio). J. Assoc. Res. Otolaryngol. 4, 219–234 (2003).
4
J. T. Corwin, D. A. Cotanche, Regeneration of sensory hair cells after acoustic trauma. Science 240, 1772–1774 (1988).
5
P. M. White, A. Doetzlhofer, Y. S. Lee, A. K. Groves, N. Segil, Mammalian cochlear supporting cells can divide and trans-differentiate into hair cells. Nature 441, 984–987 (2006).
6
L.-D. Zhao et al., Effects of DAPT and Atoh1 overexpression on hair cell production and hair bundle orientation in cultured Organ of Corti from neonatal rats. PloS One 6, e23729 (2011).
7
Z. Liu et al., Age-dependent in vivo conversion of mouse cochlear pillar and deiters’ cells to immature hair cells by Atoh1 ectopic expression. J. Neurosci. 32, 6600–6610 (2012).
8
J. C. Maass et al., Changes in the regulation of the Notch signaling pathway are temporally correlated with regenerative failure in the mouse cochlea. Front. Cell. Neurosci. 9, 110 (2015).
9
P. Chen, N. Segil, p27(Kip1) links cell proliferation to morphogenesis in the developing organ of Corti. Development 126, 1581–1590 (1999).
10
P. Chen, J. E. Johnson, H. Y. Zoghbi, N. Segil, The role of Math1 in inner ear development: Uncoupling the establishment of the sensory primordium from hair cell fate determination. Development 129, 2495–2505 (2002).
11
T. Cai, M. L. Seymour, H. Zhang, F. A. Pereira, A. K. Groves, Conditional deletion of Atoh1 reveals distinct critical periods for survival and function of hair cells in the organ of corti. J. Neurosci. 33, 10110–10122 (2013).
12
C. Woods, M. Montcouquiol, M. W. Kelley, Math1 regulates development of the sensory epithelium in the mammalian cochlea. Nat. Neurosci. 7, 1310–1318 (2004).
13
P. J. Lanford et al., Notch signalling pathway mediates hair cell development in mammalian cochlea. Nat. Genet. 21, 289–292 (1999).
14
A. E. Kiernan, R. Cordes, R. Kopan, A. Gossler, T. Gridley, The Notch ligands DLL1 and JAG2 act synergistically to regulate hair cell development in the mammalian inner ear. Development 132, 4353–4362 (2005).
15
S. T. Sinkkonen et al., Intrinsic regenerative potential of murine cochlear supporting cells. Sci. Rep. 1, 26 (2011).
16
F. Shi, J. S. Kempfle, A. S. B. Edge, Wnt-responsive Lgr5-expressing stem cells are hair cell progenitors in the cochlea. J. Neurosci. 32, 9639–9648 (2012).
17
L. Tao et al., Enhancer decommissioning imposes an epigenetic barrier to sensory hair cell regeneration. Dev. Cell 56, 2471–2485.e5 (2021).
18
A. Soufi, G. Donahue, K. S. Zaret, Facilitators and impediments of the pluripotency reprogramming factors’ initial engagement with the genome. Cell 151, 994–1004 (2012).
19
J. S. Becker, D. Nicetto, K. S. Zaret, H3K9me3-dependent heterochromatin: Barrier to cell fate changes. Trends Genet. 32, 29–41 (2016).
20
M. J. Ziller et al., Charting a dynamic DNA methylation landscape of the human genome. Nature 500, 477–481 (2013).
21
E. J. Richards, S. C. R. Elgin, Epigenetic codes for heterochromatin formation and silencing: Rounding up the usual suspects. Cell 108, 489–500 (2002).
22
K. L. Sheaffer et al., DNA methylation is required for the control of stem cell differentiation in the small intestine. Genes Dev. 28, 652–664 (2014).
23
F. Mohn et al., Lineage-specific polycomb targets and de novo DNA methylation define restriction and potential of neuronal progenitors. Mol. Cell 30, 755–766 (2008).
24
S. Domcke et al., Competition between DNA methylation and transcription factors determines binding of NRF1. Nature 528, 575–579 (2015).
25
E. C. Driver, L. Sillers, T. M. Coate, M. F. Rose, M. W. Kelley, The Atoh1-lineage gives rise to hair cells and supporting cells within the mammalian cochlea. Dev. Biol. 376, 86–98 (2013).
26
S. H. Cross, A. P. Bird, CpG islands and genes. Curr. Opin. Genet. Dev. 5, 309–314 (1995).
27
L. R. Nassar et al., The UCSC Genome Browser database: 2023 update. Nucleic Acids Res. 51, D1188–D1195 (2022).
28
A. Durand et al., Functional intestinal stem cells after Paneth cell ablation induced by the loss of transcription factor Math1 (Atoh1). Proc. Natl. Acad. Sci. U.S.A. 109, 8965–8970 (2012).
29
B. Z. Stanger, R. Datar, L. C. Murtaugh, D. A. Melton, Direct regulation of intestinal fate by Notch. Proc. Natl. Acad. Sci. U.S.A. 102, 12443–12448 (2005).
30
U. Jadhav et al., Extensive recovery of embryonic enhancer and gene memory stored in hypomethylated enhancer DNA. Mol. Cell 74, 542–554.e5 (2019).
31
M. Monk, R. L. Adams, A. Rinaldi, Decrease in DNA methylase activity during preimplantation development in the mouse. Dev. Camb. Engl. 112, 189–192 (1991).
32
M. Tahiliani et al., Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324, 930–935 (2009).
33
H. V. Yu et al., POU4F3 pioneer activity enables ATOH1 to drive diverse mechanoreceptor differentiation through a feed-forward epigenetic mechanism. Proc. Natl. Acad. Sci. U.S.A. 118, e2105137118 (2021).
34
H. S. Kaya-Okur et al., CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat. Commun. 10, 1–10 (2019).
35
C. Y. McLean et al., GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).
36
N. Yamamoto et al., Inhibition of Notch/RBP-J signaling induces hair cell formation in neonate mouse cochleas. J. Mol. Med. 84, 37–45 (2006).
37
R. G. Cavalcante, M. A. Sartor, annotatr: Genomic regions in context. Bioinform. Oxf. Engl. 33, 2381–2383 (2017).
38
W. Xie et al., Epigenomic analysis of multilineage differentiation of human embryonic stem cells. Cell 153, 1134–1148 (2013).
39
J.-S. Lee, E. Smith, A. Shilatifard, The language of histone crosstalk. Cell 142, 682–685 (2010).
40
B. E. Bernstein et al., A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).
41
Z. Chen, Q. Yin, A. Inoue, C. Zhang, Y. Zhang, Allelic H3K27me3 to allelic DNA methylation switch maintains noncanonical imprinting in extraembryonic cells. Sci. Adv. 5, eaay7246 (2019).
42
J. P. Reddington et al., Redistribution of H3K27me3 upon DNA hypomethylation results in de-repression of Polycomb target genes. Genome Biol. 14, R25 (2013).
43
S. Henikoff, J. G. Henikoff, H. S. Kaya-Okur, K. Ahmad, Efficient chromatin accessibility mapping in situ by nucleosome-tethered tagmentation. eLife 9, e63274 (2020).
44
S. Korrapati, I. Roux, E. Glowatzki, A. Doetzlhofer, Notch signaling limits supporting cell plasticity in the hair cell-damaged early postnatal murine cochlea. PLOS One 8, e73276 (2013).
45
B. C. Cox et al., Spontaneous hair cell regeneration in the neonatal mouse cochlea in vivo. Dev. Camb. Engl. 141, 816–829 (2014).
46
F. Semerci et al., Lunatic fringe-mediated Notch signaling regulates adult hippocampal neural stem cell maintenance. eLife 6, e24660 (2019).
47
L. Madisen et al., A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).
48
M. F. Rose et al., Math1 is essential for the development of hindbrain neurons critical for perinatal breathing. Neuron 64, 341–354 (2009).
49
E. Hodges et al., Directional DNA methylation changes and complex intermediate states accompany lineage specificity in the adult hematopoietic compartment. Mol. Cell 44, 17–28 (2011).
50
F. Zhang, J. H. Pomerantz, G. Sen, A. T. Palermo, H. M. Blau, Active tissue-specific DNA demethylation conferred by somatic cell nuclei in stable heterokaryons. Proc. Natl. Acad. Sci. U.S.A. 104, 4395–4400 (2007).
51
K. D. Rasmussen, K. Helin, Role of TET enzymes in DNA methylation, development, and cancer. Genes Dev. 30, 733–750 (2016).
52
G. N. L. Chua et al., Cytosine-based TET enzyme inhibitors. ACS Med. Chem. Lett. 10, 180–185 (2019).
53
M. R. Rountree, E. U. Selker, DNA methylation and the formation of heterochromatin in Neurospora crassa. Heredity 105, 38–44 (2010).
54
A. A. Iyer et al., Cellular reprogramming with ATOH1, GFI1, and POU4F3 implicate epigenetic changes and cell-cell signaling as obstacles to hair cell regeneration in mature mammals. eLife 11, e79712 (2022).
55
Y. Hao et al., Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).
56
G. Wan, G. Corfas, J. S. Stone, Inner ear supporting cells: Rethinking the silent majority. Semin. Cell Dev. Biol. 24, 448–459 (2013).
57
T. Stuart, A. Srivastava, C. Lareau, R. Satija, Multimodal single-cell chromatin analysis with Signac. bioRxiv [Preprint] (2020). https://doi.org/10.1101/2020.11.09.373613 (Accessed 10 November 2020).
58
J. C. Maass et al., Transcriptomic analysis of mouse cochlear supporting cell maturation reveals large-scale changes in notch responsiveness prior to the onset of hearing. PLoS One 11, e0167286 (2016).
59
Z. P. Stojanova, T. Kwan, N. Segil, Epigenetic regulation of Atoh1 guides hair cell development in the mammalian cochlea. Development 142, 3529–3536 (2015).
60
K. Kim et al., Epigenetic memory in induced pluripotent stem cells. Nature 467, 285–290 (2010).
61
J. M. Polo et al., Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nat. Biotechnol. 28, 848–855 (2010).
62
Y. Yin et al., Impact of cytosine methylation on DNA binding specificities of human transcription factors. Science 356, eaaj2239 (2017).
63
D. Buitrago et al., Impact of DNA methylation on 3D genome structure. Nat. Commun. 12, 3243 (2021).
64
C. Ruedl, S. Jung, DTR-mediated conditional cell ablation—Progress and challenges. Eur. J. Immunol. 48, 1114–1119 (2018).
65
P. S. Steyger, Mechanisms involved in ototoxicity. Semin. Hear. 32, 217–228 (2011).
66
D. Ding et al., Some ototoxic drugs destroy cochlear support cells before damaging sensory hair cells. Neurotox. Res. 37, 743–752 (2020).
67
A. Sriraman, T. K. Debnath, B. Xhemalce, K. M. Miller, Making it or breaking it: DNA methylation and genome integrity. Essays Biochem. 64, 687–703 (2020).
68
F. Gong, K. M. Miller, Histone methylation and the DNA damage response. Mutat. Res. 780, 37–47 (2019).
69
V. J. Wulff, M. Freshman, Age-related reduction of the RNA content of rat cardiac muscle and cerebellum. Arch. Biochem. Biophys. 95, 181–182 (1961).
70
M. J. Zhang, A. O. Pisco, S. Darmanis, J. Zou, Mouse aging cell atlas analysis reveals global and cell type-specific aging signatures. eLife 10, e62293 (2021).
71
L. Menendez et al., Generation of inner ear hair cells by direct lineage conversion of primary somatic cells. eLife 9, e55249 (2020).
72
M. A. Esteban et al., Vitamin C enhances the generation of mouse and human induced pluripotent stem cells. Cell Stem Cell 6, 71–79 (2010).
73
J. Chen et al., H3K9 methylation is a barrier during somatic cell reprogramming into iPSCs. Nat. Genet. 45, 34–42 (2013).
74
H. Marks et al., The transcriptional and epigenomic foundations of ground state pluripotency. Cell 149, 590–604 (2012).
75
A. K. Groves, Single cell transcriptomic analysis of P8 and P15 mouse cochlea (control and three overexpression conditions) and simultaneous single cell transcriptomic and accessible chromatin analysis of P1 and P8 mouse cochlea (wildtype). https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE182202. Deposited 5 February 2023.
76
J. D. Nguyen, DNA methylation in the mouse cochlea promotes maturation of supporting cells and contributes to the failure of hair cell regeneration. GEO. https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE224563. Deposited 5 February 2023.
Information & Authors
Information
Published in
Classifications
Copyright
Copyright © 2023 the Author(s). Published by PNAS. This article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).
Data, Materials, and Software Availability
Submission history
Received: January 20, 2023
Accepted: July 11, 2023
Published online: August 7, 2023
Published in issue: August 15, 2023
Keywords
Acknowledgments
We thank Welly Makmura for excellent technical assistance and Francis James for helping set up NGS alignment pipelines and providing auxiliary bioinformatics analysis. We thank past and present Lab members Ksenia Gnedeva, Litao Tao, Robert Rainey, Haoze Yu, Xizi Wang, Louise Menendez, Talon Trecek, Tuo Shi, and Jennifer Rubio for scientific discussions, Veronica Ortiz for providing initial guidance on whole-genome bisulfite sequencing (WGBS) and George Ye and Unmesh Jadhav for providing their published WGBS dataset from intestinal tissue. We are also very grateful for assistance from Jeffrey Boyd and Bernadette Masinsin of the Broad Center Flow Cytometry Facility, Seth Ruffins of the Broad Center Optical Imaging Facility, and David Ruble of the Genomics Core at Children’s Hospital Los Angeles Center for Personalized Medicine. This work was supported by F31 DC018703 to J.D.N., T32 HD060549 (supported J.D.N.)., RO1 DC014832 to A.K.G., RO1 DC015829 to N.S., and a Hearing Restoration Project Consortium award from the Hearing Health Foundation to A.K.G. and N.S.
Author contributions
J.D.N., J.L., J.G.C., and N.S. designed research; J.D.N., J.L., and T.S. performed research; J.D.N. and J.L. contributed new reagents/analytic tools; J.D.N., T.S., A.K.G., and N.S. analyzed data; and J.D.N., A.K.G., and N.S. wrote the paper.
Competing interests
The authors declare no competing interest.
Notes
This article is a PNAS Direct Submission.
Authors
Metrics & Citations
Metrics
Altmetrics
Citations
Cite this article
DNA methylation in the mouse cochlea promotes maturation of supporting cells and contributes to the failure of hair cell regeneration, Proc. Natl. Acad. Sci. U.S.A.
120 (33) e2300839120,
https://doi.org/10.1073/pnas.2300839120
(2023).
Copied!
Copying failed.
Export the article citation data by selecting a format from the list below and clicking Export.
Cited by
Loading...
View Options
View options
PDF format
Download this article as a PDF file
DOWNLOAD PDFLogin options
Check if you have access through your login credentials or your institution to get full access on this article.
Personal login Institutional LoginRecommend to a librarian
Recommend PNAS to a LibrarianPurchase options
Purchase this article to access the full text.