Differential regulation of hair cell actin cytoskeleton mediated by SRF and MRTFB

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife assessment

    This valuable study provides solid evidence implicating two transcription factors in the development of the actin cytoskeleton that shapes the mechanosensory hair bundles of the inner ear's hair cells. The work will be of interest to biologists interested in the development and maintenance of the hair bundle, both normal or impaired. Its impact would be improved by providing a mechanistic model for the observed effects.

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

The MRTF–SRF pathway has been extensively studied for its crucial role in driving the expression of a large number of genes involved in actin cytoskeleton of various cell types. However, the specific contribution of MRTF–SRF in hair cells remains unknown. In this study, we showed that hair cell-specific deletion of Srf or Mrtfb , but not Mrtf a, leads to similar defects in the development of stereocilia dimensions and the maintenance of cuticular plate integrity. We used fluorescence-activated cell sorting-based hair cell RNA-Seq analysis to investigate the mechanistic underpinnings of the changes observed in Srf and Mrtfb mutants, respectively. Interestingly, the transcriptome analysis revealed distinct profiles of genes regulated by Srf and Mrtfb , suggesting different transcriptional regulation mechanisms of actin cytoskeleton activities mediated by Srf and Mrtfb . Exogenous delivery of calponin 2 using Adeno-associated virus transduction in Srf mutants partially rescued the impairments of stereocilia dimensions and the F-actin intensity of cuticular plate, suggesting the involvement of Cnn2 , as an Srf downstream target, in regulating the hair bundle morphology and cuticular plate actin cytoskeleton organization. Our study uncovers, for the first time, the unexpected differential transcriptional regulation of actin cytoskeleton mediated by Srf and Mrtfb in hair cells, and also demonstrates the critical role of SRF–CNN2 in modulating actin dynamics of the stereocilia and cuticular plate, providing new insights into the molecular mechanism underlying hair cell development and maintenance.

Article activity feed

  1. Author Response

    Reviewer #1 (Public Review):

    This manuscript provides a comprehensive investigation of the effects of the genetic ablation of three different transcription factors (Srf, Mrtfa, and Mrtfb) in the inner ear hair cells. Based on the published data, the authors hypothesized that these transcription factors may be involved in the regulation of the genes essential for building the actin-rich structures at the apex of hair cells, the mechanosensory stereocilia and their mechanical support - the cuticular plate. Indeed, the authors found that two of these transcription factors (Srf and Mrtfb) are essential for the proper formation and/or maintenance of these structures in the auditory hair cells. Surprisingly, Srf- and Mrtfb- deficient hair cells exhibited somewhat similar abnormalities in the stereocilia and in the cuticular plates even though these transcription factors have very different effects on the hair cell transcriptome. Another interesting finding of this study is that the hair cell abnormalities in Srfdeficient mice could be rescued by AAV-mediated delivery of Cnn2, one of the downstream targets of Srf. However, despite a rather comprehensive assessment of the novel mouse models, the authors do not have yet any experimentally testable mechanistic model of how exactly Srf and Mrtfb contribute to the formation of actin cytoskeleton in the hair cells. The lack of any specific working model linking Srf and/or Mrtfb with stereocilia formation decreases the potential impact of this study.

    Major comments:

    Figures 1 & 3: The conclusion on abnormalities in the actin meshwork of the cuticular plate was based largely on the comparison of the intensities of phalloidin staining in separate samples from different groups. In general, any comparison of the intensity of fluorescence between different samples is unreliable, no matter how carefully one could try matching sample preparation and imaging conditions. In this case, two other techniques would be more convincing: 1) quantification of the volume of the cuticular plates from fluorescent images; and 2) direct examination of the cuticular plates by transmission electron microscopy (TEM).

    In fact, the manuscript provides no single TEM image of the F-actin abnormalities either in the cuticular plate or in the stereocilia, even though these abnormalities seem to be the major focus of the study. Overall, it is still unclear what exactly Srf or Mrtfb deficiencies do with F-actin in the hair cells.

    Yes, we agree. As suggested by the reviewer, to directly examine the defects in F-actin organization within the cuticular plate of mutant mice, we conducted Transmission Electron Microscopy (TEM) analyses. The results, as presented in the revised Figures 1 and 4 (panels F, G, and E, F, respectively), provide crucial insights into the structural changes in the cuticular plate. Meanwhile, the comparison of the volume of the phalloidin labeled cuticular plate after 3-D reconstruction using Imaris software was conducted and shown in Author response image 1. The results of the cuticular plate (CP) volume were consistent with the relative F-actin intensity change of the cuticular plate in the revised Figures 1B and 4B. For the TEM analysis of the stereocilia, we regret that due to time constraints, we were unable to collect TEM images of stereocilia with sufficient quality for a meaningful comparison. However, we believe that the data we have presented sufficiently addresses the primary concerns, and we appreciate the reviewers’ understanding of these limitations.

    Author response image 1.

    Figures 2 & 4 represent another example of how deceiving could be a simple comparison of the intensity of fluorescence between the genotypes. It is not clear whether the reduced immunofluorescence of the investigated molecules (ESPN1, EPS8, GNAI3, or FSCN2) results from their mis-localization or represents a simple consequence of the fact that a thinner stereocilium would always have a smaller signal of the protein of interest, even though the ratio of this protein to the number of actin filaments remains unchanged. According to my examination of the representative images of these figures, loss of Srf produces mis-localization of the investigated proteins and irregular labeling in different stereocilia of the same bundle, while loss of Mrtfb does not. Obviously, a simple quantification of the intensity of fluorescence conceals these important differences.

    Yes, we agree. In addition to the quantification of tip protein intensity, we have added a few more analyses in the revised Figure 3 and Figure 6, such as the percentage of row 1 tip stereocilia with tip protein staining and the percentage of IHCs with tip protein staining on row 2 tip. Using the results mentioned above, the differences in the expression level, the row-specific distribution and the irregular labeling of tip proteins between the control and the mutants can be analyzed more thoroughly.

    Reviewer #2 (Public Review):

    The analysis of bundle morphology using both confocal and SEM imaging is a strength of the paper and the authors have some nice images, especially with SEM. Still, the main weakness is that it is unclear how significant their findings are in terms of understanding bundle development; the mouse phenotypes are not distinct enough to make it clear that they serve different functions so the reader is left wondering what the main takeaway is.

    Based on the reviewer’s comments, in this revised manuscript, we put more emphasis on describing the effects of SRF and MRTFB on key tip proteins’ localization pattern during stereocilia development, represented by ESPN1, EPS8 and GNAI3, as well as the effects of SRF and MRTFB on the F-actin organization of cuticular plate using TEM. We have made substantial efforts to interpret the mechanistic underpinnings of the roles of SRF and MRTFB in hair cells. This is reflected in the revised Figures 1, 3, 4, 6, and 10, where we provide more comprehensive insights into the mechanisms at play.

    We interpret our data in a way that both SRF and MRTF regulate the development and maintenance of the hair cell’s actin cytoskeleton in a complementary manner. Deletion of either gene thus results in somewhat similar phenotypes in hair cell morphology, despite the surprising lack of overlap of SRF and MRTFB downstream targets in the hair cell.

    In Figure 1 and 3, changes in bundle morphology clearly don't occur until after P5. Widening still occurs to some extent but lengthening does not and instead the stereocilia appear to shrink in length. EPS8 levels appear to be the most reduced of all the tip proteins (Srf mutants) so I wonder if these mutants are just similar to an EPS8 KO if the loss of EPS8 occurred postnatally (P0-P5).

    To address this question, we performed EPS8 staining on the control and Srf cKO hair cells at P4 and P10. We found that the dramatic decrease of the row 1 tip signal for EPS8 started since P4 in Srf cKO IHCs. Although the major hair bundle phenotype of Eps8 KO, including the defects of row 1 stereocilia lengthening and additional rows of short stereocilia also appeared in Srf cKO IHCs, there are still some bundle morphology differences between Eps8 KO and Srf cKO. For example, firstly, both Eps8 KO OHCs and IHCs showed additional rows of short stereocilia, but we only observed additional rows of short stereocilia in Srf cKO IHCs. Secondly, in Valeria Zampini’s study, SEM and TEM images did not show an obvious reduction of row 2 stereocilia widening (P18-P35), while our analysis of SEM images confirmed that the width of row 2 IHC stereocilia was drastically reduced by 40% in Srf cKO (P15). Generally, we think although Srf cKO hair bundles are somewhat similar to Eps8 KO, the Srf cKO hair bundle phenotype might be governed by multiple candidate genes cooperatively.

    Reference:

    Valeria Zampini, et al. Eps8 regulates hair bundle length and functional maturation of mammalian auditory hair cells. PLoS Biol. 2011 Apr;9(4): e1001048.

    A major shortcoming is that there are few details on how the image analyses were done. Were SEM images corrected for shrinkage? How was each of the immunocytochemistry quantitation (e.g., cuticular plates for phalloidin and tip staining for antibodies) done? There are multiple ways of doing this but there are few indications in the manuscript.

    We apologize for not making the description of the procedure of images analyses clear enough. As described in Nicolas Grillet group’s study, live and mildly-fixed IHC stereocilia have similar dimensions, while SEM preparation results in a hair bundle at a 2:3 scale compared to the live preparation. In our study, the hair cells selected for SEM imaging and measurements were located in the basal turn (30-32kHz), while the hair cells selected for fluorescence-based imaging and measurements were located in the middle turn (20-24kHz) or the basal turn (32-36kHz). Although our SEM imaging and fluorescence-based imaging of basal turn’s hair bundles were not from the same area exactly, the control hair bundles with SEM imaging have reduced row 1 stereocilia length by 10%-20%, compared to the control hair bundles with fluorescence-based imaging (revised Figure 2 and Figure 5). Generally, our stereocilia dimensions data showed appropriate shrinkage caused by the SEM preparation.

    Recognizing the need for clarity, we have provided a detailed description of our image quantification and analysis procedures in the “Materials and Methods” section, specifically under “Immunocytochemistry.” This will aid readers in understanding our methodologies and ensure transparency in our approach.

    Reference:

    Katharine K Miller, et al. Dimensions of a Living Cochlear Hair Bundle. Front Cell Dev Biol. 2021 Nov 25:9:742529.

    The tip protein analysis in Figs 2 and 4 is nice but it would be nice for the authors to show the protein staining separately from the phalloidin so you could see how restricted to the tips it is (each in grayscale). This is especially true for the CNN2 labeling in Fig 7 as it does not look particularly tip specific in the x-y panels. It would be especially important to see the antibody staining in the reslices separate from phalloidin.

    Thank you for the suggestions. We have shown tip proteins staining in grayscale separately from the phalloidin in the revised Figure 3 and Figure 6. To clearly show the tip-specific localization of CNN2, we conducted CNN2 staining at different ages during hair bundle development and showed CNN2 labeling in grayscale and in reslices in revised Figure 9-figure supplement 1B.

    In Fig 6, why was the transcriptome analysis at P2 given that the phenotype in these mice occurs much later? While redoing the transcriptome analysis is probably not an option, an alternative would be to show more examples of EPS8/GNAI/CNN2 staining in the KO, but at younger ages closer to the time of PCR analysis, such as at P5. Pinpointing when the tip protein intensities start to decrease in the KOs would be useful rather than just showing one age (P10).

    We agree with the reviewer. To address this question, we have performed ESPN1, EPS8 and GNAI3 staining on the control and the mutant’s hair cells at P4, P10 and P15 (the revised Figures 3 and 6). According to the new results, we found that the dramatic decreases of the row 1 tip signal for ESPN1 and EPS8 started since P4 in Srf cKO IHCs, is consistent with the appearance of the mild reduction of row 1 stereocilia length in P5 Srf cKO IHCs. For Mrtfb cKO hair cells, the obvious reduction of the row 1 tip signal for ESPN1 was observed until P10. However, a few genes related to cell adhesion and regulation of actin cytoskeleton were significantly down-regulated in P2 Mrtfb deficient hair cell transcriptome. We think that in hair cells the MRTFB may not play a major role in the regulation of stereocilia development, so the morphological defects of stereocilia happened much later in the Mrtfb mutant than in the Srf mutant.

    While it is certainly interesting if it turns out CNN2 is indeed at tips in this phase, the experiments do not tell us that much about what role CNN2 may be playing. It is notable that in Fig 7E in the control+GFP panel, CNN2 does not appear to be at the tips. Those images are at P11 whereas the images in panel A are at P6 so perhaps CNN2 decreases after the widening phase. An important missing control is the Anc80L65-Cnn2 AAV in a wild-type cochlea.

    We agree with the reviewer. We have conducted more immunostaining experiments to confirm the expression pattern of CNN2 during the stereocilia development, from P0 to P11. The results were included in the revised Figure 9-figure supplement 1B. As the reviewer suggested, CNN2 expression pattern in control cochlea injected with Anc80L65-Cnn2 AAV has also been provided in revised Figure 9E.

  2. eLife assessment

    This valuable study provides solid evidence implicating two transcription factors in the development of the actin cytoskeleton that shapes the mechanosensory hair bundles of the inner ear's hair cells. The work will be of interest to biologists interested in the development and maintenance of the hair bundle, both normal or impaired. Its impact would be improved by providing a mechanistic model for the observed effects.

  3. Reviewer #1 (Public Review):

    This manuscript provides a comprehensive investigation of the effects of the genetic ablation of three different transcription factors (Srf, Mrtfa, and Mrtfb) in the inner ear hair cells. Based on the published data, the authors hypothesized that these transcription factors may be involved in the regulation of the genes essential for building the actin-rich structures at the apex of hair cells, the mechanosensory stereocilia and their mechanical support - the cuticular plate. Indeed, the authors found that two of these transcription factors (Srf and Mrtfb) are essential for the proper formation and/or maintenance of these structures in the auditory hair cells. Surprisingly, Srf- and Mrtfb- deficient hair cells exhibited somewhat similar abnormalities in the stereocilia and in the cuticular plates even though these transcription factors have very different effects on the hair cell transcriptome. Another interesting finding of this study is that the hair cell abnormalities in Srf-deficient mice could be rescued by AAV-mediated delivery of Cnn2, one of the downstream targets of Srf. However, despite a rather comprehensive assessment of the novel mouse models, the authors do not have yet any experimentally testable mechanistic model of how exactly Srf and Mrtfb contribute to the formation of actin cytoskeleton in the hair cells. The lack of any specific working model linking Srf and/or Mrtfb with stereocilia formation decreases the potential impact of this study.

    Major comments:

    Figures 1 & 3: The conclusion on abnormalities in the actin meshwork of the cuticular plate was based largely on the comparison of the intensities of phalloidin staining in separate samples from different groups. In general, any comparison of the intensity of fluorescence between different samples is unreliable, no matter how carefully one could try matching sample preparation and imaging conditions. In this case, two other techniques would be more convincing: 1) quantification of the volume of the cuticular plates from fluorescent images; and 2) direct examination of the cuticular plates by transmission electron microscopy (TEM).

    In fact, the manuscript provides no single TEM image of the F-actin abnormalities either in the cuticular plate or in the stereocilia, even though these abnormalities seem to be the major focus of the study. Overall, it is still unclear what exactly Srf or Mrtfb deficiencies do with F-actin in the hair cells.

    Figures 2 & 4 represent another example of how deceiving could be a simple comparison of the intensity of fluorescence between the genotypes. It is not clear whether the reduced immunofluorescence of the investigated molecules (ESPN1, EPS8, GNAI3, or FSCN2) results from their mis-localization or represents a simple consequence of the fact that a thinner stereocilium would always have a smaller signal of the protein of interest, even though the ratio of this protein to the number of actin filaments remains unchanged. According to my examination of the representative images of these figures, loss of Srf produces mis-localization of the investigated proteins and irregular labeling in different stereocilia of the same bundle, while loss of Mrtfb does not. Obviously, a simple quantification of the intensity of fluorescence conceals these important differences.

  4. Reviewer #2 (Public Review):

    The analysis of bundle morphology using both confocal and SEM imaging is a strength of the paper and the authors have some nice images, especially with SEM. Still, the main weakness is that it is unclear how significant their findings are in terms of understanding bundle development; the mouse phenotypes are not distinct enough to make it clear that they serve different functions so the reader is left wondering what the main takeaway is.

    In Figure 1 and 3, changes in bundle morphology clearly don't occur until after P5. Widening still occurs to some extent but lengthening does not and instead the stereocilia appear to shrink in length. EPS8 levels appear to be the most reduced of all the tip proteins (Srf mutants) so I wonder if these mutants are just similar to an EPS8 KO if the loss of EPS8 occurred postnatally (P0-P5).

    A major shortcoming is that there are few details on how the image analyses were done. Were SEM images corrected for shrinkage? How was each of the immunocytochemistry quantitation (e.g., cuticular plates for phalloidin and tip staining for antibodies) done? There are multiple ways of doing this but there are few indications in the manuscript.

    The tip protein analysis in Figs 2 and 4 is nice but it would be nice for the authors to show the protein staining separately from the phalloidin so you could see how restricted to the tips it is (each in grayscale). This is especially true for the CNN2 labeling in Fig 7 as it does not look particularly tip specific in the x-y panels. It would be especially important to see the antibody staining in the reslices separate from phalloidin.

    In Fig 6, why was the transcriptome analysis at P2 given that the phenotype in these mice occurs much later? While redoing the transcriptome analysis is probably not an option, an alternative would be to show more examples of EPS8/GNAI/CNN2 staining in the KO, but at younger ages closer to the time of PCR analysis, such as at P5. Pinpointing when the tip protein intensities start to decrease in the KOs would be useful rather than just showing one age (P10).

    While it is certainly interesting if it turns out CNN2 is indeed at tips in this phase, the experiments do not tell us that much about what role CNN2 may be playing. It is notable that in Fig 7E in the control+GFP panel, CNN2 does not appear to be at the tips. Those images are at P11 whereas the images in panel A are at P6 so perhaps CNN2 decreases after the widening phase. An important missing control is the Anc80L65-Cnn2 AAV in a wild-type cochlea.