Müller glia cell cycle re-activation by simultaneous cyclin D1 overexpression and p27kip1 knockdown promotes retinal regeneration in mice

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife assessment

    The manuscript presents a potentially important strategy to stimulate mammalian Müller glia to proliferate in vivo by manipulating cell cycle components. The findings are likely to appeal to retinal specialists and neuroscientists in general. However, the evidence that these cells become neurogenic is lacking/incomplete, suggesting that additional barriers exist to stimulate the regeneration of retinal neurons.

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

Harnessing the regenerative potential of endogenous stem cells to restore lost neurons is a promising strategy for treating neurodegenerative disorders. Müller glia (MG), the primary glial cell type in the retina, exhibit remarkable regenerative abilities in lower vertebrate species, such as zebrafish and amphibians, where injury induces MG to proliferate and differentiate into various retinal neuron types. The regenerative potential of mammalian MG is constrained by their inherent inability to re-enter the cell cycle, likely due to high levels of the cell cycle inhibitor p27 Kip1 and low levels of cyclin D1 observed in adult mouse MG. In this study, we found that adeno-associated virus (AAV)-mediated cyclin D1 overexpression and p27 Kip1 knockdown exerts a strong synergistic effect on MG proliferation. MG proliferation induced by this treatment was potent but self-limiting, as MG did not undergo uncontrolled proliferation or lead to retinal neoplasia. Single-cell RNA sequencing (scRNA-seq) revealed that cell cycle reactivation leads to immunosuppression and dedifferentiation of MG. Notably, scRNA-seq analysis identified a new cluster of rod-like MG cells expressing both rod and MG genes, which was further validated by RNA in situ hybridization. Cell cycle reactivation also led to de novo genesis of bipolar- and amacrine-like cells from MG. Overall, our findings suggest that AAV- mediated cyclin D1 overexpression and p27 Kip1 knockdown stimulate MG proliferation and promote MG reprogramming. This approach may be a promising strategy, especially when combined with other regeneration-promoting factors, to enhance MG-mediated retinal repair.

Article activity feed

  1. eLife assessment

    The manuscript presents a potentially important strategy to stimulate mammalian Müller glia to proliferate in vivo by manipulating cell cycle components. The findings are likely to appeal to retinal specialists and neuroscientists in general. However, the evidence that these cells become neurogenic is lacking/incomplete, suggesting that additional barriers exist to stimulate the regeneration of retinal neurons.

  2. Reviewer #1 (Public Review):

    Summary:

    In this manuscript, Wu et al. introduce a novel approach to reactivate the Muller glia cell cycle in the mouse retina by simultaneously reducing p27Kip1 and increasing cyclin D1 using a single AAV vector. The approach effectively promotes Muller glia proliferation and reprograming without disrupting retinal structure or function. Interestingly, reactivation of the Muller glia cell cycle downregulates IFN pathway, which may contribute to the induced retinal regeneration. The results presented in this manuscript may offer a promising approach for developing Müller glia cell-mediated regenerative therapies for retinal diseases.

    Strengths:

    The data are convincing and supported by appropriate, validated methodology. These results are both technically and scientifically exciting and are likely to appeal to retinal specialists and neuroscientists in general.

    Weaknesses:

    There are some data gaps that need to be addressed.

    (1) Please label the time points of AAV injection, EdU labeling, and harvest in Figure 1B.

    (2) What fraction of Müller cells were transduced by AAV under the experimental conditions?

    (3) It seems unusually rapid for MG proliferation to begin as early as the third day after CCA injection. Can the authors provide evidence for cyclin D1 overexpression and p27 Kip1 knockdown three days after CCA injection?

    (4) The authors reported that MG proliferation largely ceased two weeks after CCA treatment. While this is an interesting finding, the explanation that it might be due to the dilution of AAV episomal genome copies in the dividing cells seems far-fetched.

  3. Reviewer #2 (Public Review):

    This manuscript by Wu, Liao et al. reports that simultaneous knockdown of P27Kip1 with overexpression of Cyclin D can stimulate Muller glia to re-enter the cell cycle in the mouse retina. There is intense interest in reprogramming mammalian muller glia into a source for neurogenic progenitors, in the hopes that these cells could be a source for neuronal replacement in neurodegenerative diseases. Previous work in the field has shown ways in which mouse Muller glia can be neurogenically reprogrammed and these studies have shown cell cycle re-entry prior to neurogenesis. In other works, typically, the extent of glial proliferation is limited, and the authors of this study highlight the importance of stimulating large numbers of Muller glia to re-enter the cell cycle with the hopes they will differentiate into neurons. While the evidence for stimulating proliferation in this study is convincing, the evidence for neurogenesis in this study is not convincing or robust, suggesting that stimulating cell cycle-reentry may not be associated with increasing regeneration without another proneural stimulus.

    Below are concerns and suggestions.

    Intro:

    (1) The authors cite past studies showing "direct conversion" of MG into neurons. However, these studies (PMID: 34686336; 36417510) show EdU+ MG-derived neurons suggesting cell cycle re-entry does occur in these strategies of proneural TF overexpression.

    (2) Multiple citations are incorrectly listed, using the authors first name only (i.e. Yumi, et al; Levi, et al;). Studies are also incompletely referenced in the references.

    Figure 1:
    (3) When are these experiments ending? On Figure 1B it says "analysis" on the end of the paradigm without an actual day associated with this. This is the case for many later figures too. The authors should update the paradigms to accurately reflect experimental end points.

    (4) Are there better representative pictures between P27kd and CyclinD OE, the EdU+ counts say there is a 3 fold increase between Figure 1D&E, however the pictures do not reflect this. In fact, most of the Edu+ cells in Figure 1E don't seem to be Sox9+ MG but rather horizontally oriented nuclei in the OPL that are likely microglia.

    (5) Is the infection efficacy of these viruses different between different combinations (i.e. CyclinD OE vs. P27kd vs. control vs. CCA combo)? As the counts are shown in Figure 1G only Sox9+/Edu+ cells are shown not divided by virus efficacy. If these are absolute counts blind to where the virus is and how many cells the virus hits, if the virus efficacy varies in efficiency this could drive absolute differences that aren't actually biological.

    (6) According to the Jax laboratories, mice aren't considered aged until they are over 18months old. While it is interesting that CCA treatment does not seem to lose efficacy over maturation I would rephrase the findings as the experiment does not test this virus in aged retinas.

    (7) Supplemental Figure 2c-d. These viruses do not hit 100% of MG, however 100% of the P27Kip staining is gone in the P27sh1 treatment, even the P27+ cell in the GCL that is likely an astrocyte has no staining in the shRNA 1 picture. Why is this?

    Figure 2
    (8) Would you expect cells to go through two rounds of cell cycle in such a short time? The treatment of giving Edu then BrdU 24 hours later would have to catch a cell going through two rounds of division in a very short amount of time. Again the end point should be added graphically to this figure.

    Figure 3
    (9) I am confused by the mixing of ratios of viruses to indicate infection success. I know mixtures of viruses containing CCA or control GFP or a control LacZ was injected. Was the idea to probe for GFP or LacZ in the single cell data to see which cells were infected but not treated? This is not shown anywhere?

    (10) The majority of glia sorted from TdTomato are probably not infected with virus. Can you subset cells that were infected only for analysis? Otherwise it makes it very hard to make population judgements like Figure 3E-H if a large portion are basically WT glia.

    (11) Figure 3C you can see Rho is expressed everywhere which is common in studies like this because the ambient RNA is so high. This makes it very hard to talk about "Rod-like" MG as this is probably an artifact from the technique. Most all scRNA-seq studies from MG-reprogramming have shown clusters of "rods" with MG hybrid gene expression and these had in the past just been considered an artifact.

    (12) It is mentioned the "glial" signature is downregulated in response to CCA treatment. Where is this shown convincingly? Figure H has a feature plot of Glul , which is not clear it is changed between treatments. Otherwise MG genes are shown as a function of cluster not treatment.

    Figure 4
    (13) The authors should be commended for being very careful in their interpretations. They employ the proper controls (Er-Cre lineage tracing/EdU-pulse chasing/scRNA-seq omics) and were very careful to attempt to see MG-derived rods. This makes the conclusion from the FISH perplexing. The few puncta dots of Rho and GNAT in MG are not convincing to this reviewer, Rho and GNAT dots are dense everywhere throughout the ONL and if you drew any random circle in the ONL it would be full of dots. The rigor of these counts also comes into question because some dots are picked up in MG in the INL even in the control case. This is confusing because baseline healthy MG do not express RNA-transcripts of these Rod genes so what is this picking up? Taken together, the conclusion that there are Rod-like MG are based off scRNA-seq data (which is likely ambient contamination) and these FISH images. I don't think this data warrants the conclusion that MG upregulate Rod genes in response to CCA.

    Figure 5
    (14) Similar point to above but this Glul probe seems odd, why is it throughout the ONL but completely dark through the IPL, this should also be in astrocytes can you see it in the GCL? These retinas look cropped at the INL where below is completely black. The whole retinal section should be shown. Antibodies exist to GS that work in mouse along with many other MG genes, IHC or western blots could be done to better serve this point.

    Figure 6
    (15) Figure 6D is not a co-labeled OTX2+/ TdTomato+ cell, Otx2 will fill out the whole nucleus as can be seen with examples from other MG-reprogramming papers in the field (Hoang, et al. 2020; Todd, et al. 2020; Palazzo, et al. 2022). You can clearly see in the example in Figure 6D the nucleus extending way beyond Otx2 expression as it is probably overlapping in space. Other examples should be shown, however, considering less than 1% of cells were putatively Otx2+, the safer interpretation is that these cells are not differentiating into neurons. At least 99.5% are not.

    (16) Same as above Figure 6I is not convincingly co-labeled HuC/D is an RNA-binding protein and unfortunately is not always the clearest stain but this looks like background haze in the INL overlapping. Other amacrine markers could be tested, but again due to the very low numbers, I think no neurogenesis is occurring.

    (17) In the text the authors are accidently referring to Figure 6 as Figure 7.

    Figure 7
    (18) I like this figure and the concept that you can have additional MG proliferating without destroying the retina or compromising vision. This is reminiscent of the chick MG reprogramming studies in which MG proliferate in large numbers and often do not differentiate into neurons yet still persist de-laminated for long time points.

    General:
    (19) The title should be changed, as I don't believe there is any convincing evidence of regeneration of neurons. Understanding the barriers to MG cell-cycle re-entry are important and I believe the authors did a good job in that respect, however it is an oversell to report regeneration of neurons from this data.

    (20) This paper uses multiple mouse lines and it is often confusing when the text and figures switch between models. I think it would be helpful to readers if the mouse strain was added to graphical paradigms in each figure when a different mouse line is employed.

  4. Author response:

    Public Reviews:

    Reviewer #1 (Public Review):

    Summary:

    In this manuscript, Wu et al. introduce a novel approach to reactivate the Muller glia cell cycle in the mouse retina by simultaneously reducing p27Kip1 and increasing cyclin D1 using a single AAV vector. The approach effectively promotes Muller glia proliferation and reprograming without disrupting retinal structure or function. Interestingly, reactivation of the Muller glia cell cycle downregulates IFN pathway, which may contribute to the induced retinal regeneration. The results presented in this manuscript may offer a promising approach for developing Müller glia cell-mediated regenerative therapies for retinal diseases.

    Strengths:

    The data are convincing and supported by appropriate, validated methodology. These results are both technically and scientifically exciting and are likely to appeal to retinal specialists and neuroscientists in general.

    Weaknesses:

    There are some data gaps that need to be addressed.

    (1) Please label the time points of AAV injection, EdU labeling, and harvest in Figure 1B.

    We thank the reviewer for highlighting the lack of clarity in our experimental design. We will label all experiment timelines in the figures where appropriate in the revised version.

    (2) What fraction of Müller cells were transduced by AAV under the experimental conditions?

    We apologize for not clearly conveying the transduction efficiency. The retinal region adjacent to the injection site, typically near the central retina, exhibits a transduction efficiency of nearly 100%. In contrast, the peripheral retina shows a lower transduction efficiency compared to the central region. We will include the quantification of AAV transduction efficiency in the revised manuscript.

    The quantification of Edu+ MG or other markers was conducted in the area with the highest efficiency.

    (3) It seems unusually rapid for MG proliferation to begin as early as the third day after CCA injection. Can the authors provide evidence for cyclin D1 overexpression and p27 Kip1 knockdown three days after CCA injection?

    In our pilot study, we tested the onset time of GFP expression from AAV-GFAP-GFP following intravitreal injection. We observed GFP expression in MG as early as two days post-infection. These findings will be included in the revised manuscript. Additionally, we plan to perform qPCR or Western blot analysis to confirm cyclin D1 overexpression and p27kip1 knockdown at the onset of Müller glia proliferation, which will also be included in the revised manuscript.

    (4) The authors reported that MG proliferation largely ceased two weeks after CCA treatment. While this is an interesting finding, the explanation that it might be due to the dilution of AAV episomal genome copies in the dividing cells seems far-fetched.

    We believe that the lack of durability in high Cyclin D1 and low p27kip1 levels in MG contributes to the cessation of their proliferation. A potential reason for the loss of high Cyclin D1 overexpression and p27kip1 knockdown during MG proliferation could be the dilution of the AAV episomal genome. However, testing this hypothesis is challenging. Instead, we plan to provide direct evidence in the revised manuscript by examining the levels of Cyclin D1 and p27kip1 in the retina treated with CCA before and after the peak of MG proliferation.

    Reviewer #2 (Public Review):

    This manuscript by Wu, Liao et al. reports that simultaneous knockdown of P27Kip1 with overexpression of Cyclin D can stimulate Muller glia to re-enter the cell cycle in the mouse retina. There is intense interest in reprogramming mammalian muller glia into a source for neurogenic progenitors, in the hopes that these cells could be a source for neuronal replacement in neurodegenerative diseases. Previous work in the field has shown ways in which mouse Muller glia can be neurogenically reprogrammed and these studies have shown cell cycle re-entry prior to neurogenesis. In other works, typically, the extent of glial proliferation is limited, and the authors of this study highlight the importance of stimulating large numbers of Muller glia to re-enter the cell cycle with the hopes they will differentiate into neurons. While the evidence for stimulating proliferation in this study is convincing, the evidence for neurogenesis in this study is not convincing or robust, suggesting that stimulating cell cycle-reentry may not be associated with increasing regeneration without another proneural stimulus.

    Below are concerns and suggestions.

    Intro:

    (1) The authors cite past studies showing "direct conversion" of MG into neurons. However, these studies (PMID: 34686336; 36417510) show EdU+ MG-derived neurons suggesting cell cycle re-entry does occur in these strategies of proneural TF overexpression.

    We thank the reviewer for pointing this out. We will revise the statement to "MG neurogenesis," which encompasses both direct conversion and Müller glia proliferation followed by neuronal differentiation.

    (2) Multiple citations are incorrectly listed, using the authors first name only (i.e. Yumi, et al; Levi, et al;). Studies are also incompletely referenced in the references.

    We apologize for the mistake with the reference. We will fix these mistakes in the revised version.

    Figure 1:

    (3) When are these experiments ending? On Figure 1B it says "analysis" on the end of the paradigm without an actual day associated with this. This is the case for many later figures too. The authors should update the paradigms to accurately reflect experimental end points.

    We thank the reviewer for highlighting the lack of clarity in our experimental design. We will label all experiment timelines in the figures where appropriate in the revised version.

    (4) Are there better representative pictures between P27kd and CyclinD OE, the EdU+ counts say there is a 3 fold increase between Figure 1D&E, however the pictures do not reflect this. In fact, most of the Edu+ cells in Figure 1E don't seem to be Sox9+ MG but rather horizontally oriented nuclei in the OPL that are likely microglia.

    Thanks to the reviewer for pointing this out. We will replace the image of Cyclin D1 which a better representative image.

    (5) Is the infection efficacy of these viruses different between different combinations (i.e. CyclinD OE vs. P27kd vs. control vs. CCA combo)? As the counts are shown in Figure 1G only Sox9+/Edu+ cells are shown not divided by virus efficacy. If these are absolute counts blind to where the virus is and how many cells the virus hits, if the virus efficacy varies in efficiency this could drive absolute differences that aren't actually biological.

    Because the AAV-GFAP-Cyclin D1 and AAV-GFAP-Cyclin D1-p27kip1 shRNA viruses do not carry a fluorescent reporter gene, we cannot easily measure viral efficacy in the same experiment. We believe that variations in viral efficacy cannot account for the significant differences in MG proliferation for two reasons: 1) We injected the same titer for all three viruses, and 2) Viral infection efficacy is very high, approaching 100% in the central retina. Nonetheless, to rule out the possibility that the differences in MG proliferation among the Cyclin D overexpression, p27kip1 knockdown, and CCA groups are due to variations in viral efficacy, we will include the p27kip1 knockdown and Cyclin D1 overexpression efficiencies for all four groups using qPCR and/or Western blot analysis in the revised manuscript.

    (6) According to the Jax laboratories, mice aren't considered aged until they are over 18months old. While it is interesting that CCA treatment does not seem to lose efficacy over maturation I would rephrase the findings as the experiment does not test this virus in aged retinas.

    Thank you to the reviewer for bringing this to our attention. We will void using “aged mice” in our revised manuscript.

    (7) Supplemental Figure 2c-d. These viruses do not hit 100% of MG, however 100% of the P27Kip staining is gone in the P27sh1 treatment, even the P27+ cell in the GCL that is likely an astrocyte has no staining in the shRNA 1 picture. Why is this?

    For Supplementary Figure 2c-d, we focused on the central area where knockdown efficiency was high, approaching 100%. We will replace this image with one that includes both high and low Müller glia transduction efficiency regions, clearly demonstrating the complete loss of p27kip1 staining in the area of high transduction efficiency.

    Figure 2

    (8) Would you expect cells to go through two rounds of cell cycle in such a short time? The treatment of giving Edu then BrdU 24 hours later would have to catch a cell going through two rounds of division in a very short amount of time. Again the end point should be added graphically to this figure.

    We thank the reviewer for raising this important point. While the typical cell cycle time for human cells is approximately 24 hours, we hypothesized that 24 hours would be the most likely timepoint to capture cells continuously progressing through the cell cycle. However, we acknowledge that we cannot exclude the possibility of some cells entering a second cell cycle at much later timepoints.

    In the revised manuscript, we will carefully qualify our conclusion to state that the majority of MG do not immediately undergo another cell division, rather than making a definitive statement. This more cautious phrasing will better reflect the limitations of the 24-hour timepoint and allow for the potential of a small subset of cells proceeding through additional rounds of division at later stages.

    Figure 3

    (9) I am confused by the mixing of ratios of viruses to indicate infection success. I know mixtures of viruses containing CCA or control GFP or a control LacZ was injected. Was the idea to probe for GFP or LacZ in the single cell data to see which cells were infected but not treated? This is not shown anywhere?

    The virus infection was not uniform across the entire retina. To mark the infection hotspots, we added 10% GFP virus to the mixture. Regions of the retina with low infection efficiency were removed by dissection and excluded from the scRNA-seq analysis. We apologize for not clearly explaining this methodological detail in the original text, and will update the Methods section accordingly.

    (10) The majority of glia sorted from TdTomato are probably not infected with virus. Can you subset cells that were infected only for analysis? Otherwise it makes it very hard to make population judgements like Figure 3E-H if a large portion are basically WT glia.

    This question is related to the last one. Since the regions with high virus infection efficiency were selectively dissected and isolated for analysis, the percentage of CCA-infected MG should constitute the majority in the scRNA-seq data.

    (11) Figure 3C you can see Rho is expressed everywhere which is common in studies like this because the ambient RNA is so high. This makes it very hard to talk about "Rod-like" MG as this is probably an artifact from the technique. Most all scRNA-seq studies from MG-reprogramming have shown clusters of "rods" with MG hybrid gene expression and these had in the past just been considered an artifact.

    We agree that the low levels of Rho in other MG clusters (such as quiescent, reactivated, and proliferating MG) are likely due to RNA contamination. However, the level of Rho in the rod-like MG is significantly higher than in the other clusters, indicating that this is unlikely to be solely due to contamination.

    As shown in Supplementary Figure 7A-C, a cluster of MG-rod hybrid cells (cluster C4) was present in all three experimental groups at similar ratios, and this hybrid cluster was excluded from further analysis. In contrast, the rod-like Müller glia (cluster C3) were predominantly found in the CCA and CCANT groups, suggesting a genuine response to CCA treatment.

    Furthermore, we will conduct Rho and Gnat1 RNA in situ hybridization on the dissociated retinal cells to further support the conclusion that rod-specific genes are upregulated in a subset of MG in the revised manuscript.

    (12) It is mentioned the "glial" signature is downregulated in response to CCA treatment. Where is this shown convincingly? Figure H has a feature plot of Glul , which is not clear it is changed between treatments. Otherwise MG genes are shown as a function of cluster not treatment.

    We will add box plots of several MG-specific genes to better illustrate the downregulation of the glial signature in the relevant cell cluster in the revised manuscript.

    Figure 4

    (13) The authors should be commended for being very careful in their interpretations. They employ the proper controls (Er-Cre lineage tracing/EdU-pulse chasing/scRNA-seq omics) and were very careful to attempt to see MG-derived rods. This makes the conclusion from the FISH perplexing. The few puncta dots of Rho and GNAT in MG are not convincing to this reviewer, Rho and GNAT dots are dense everywhere throughout the ONL and if you drew any random circle in the ONL it would be full of dots. The rigor of these counts also comes into question because some dots are picked up in MG in the INL even in the control case. This is confusing because baseline healthy MG do not express RNA-transcripts of these Rod genes so what is this picking up? Taken together, the conclusion that there are Rod-like MG are based off scRNA-seq data (which is likely ambient contamination) and these FISH images. I don't think this data warrants the conclusion that MG upregulate Rod genes in response to CCA.

    We performed RNA in situ hybridization on retinal sections because we aimed to correlate cell localization with rod gene expression. We understand the reviewer’s concern that the punctate signals of Rho and GNAT1 in the ONL MG may actually originate from neighboring rods. In the revised manuscript, we will conduct RNAscope on dissociated retinal cells to avoid this issue.

    Figure 5

    (14) Similar point to above but this Glul probe seems odd, why is it throughout the ONL but completely dark through the IPL, this should also be in astrocytes can you see it in the GCL? These retinas look cropped at the INL where below is completely black. The whole retinal section should be shown. Antibodies exist to GS that work in mouse along with many other MG genes, IHC or western blots could be done to better serve this point.

    Indeed, the GCL was cropped out in Figure 5 A-B. We have other images with all retinal layers, which we will use in the revised manuscript. Additionally, we will perform the GS antibody staining to demonstrate partial MG dedifferentiation following CCA treatment.

    Figure 6

    (15) Figure 6D is not a co-labeled OTX2+/ TdTomato+ cell, Otx2 will fill out the whole nucleus as can be seen with examples from other MG-reprogramming papers in the field (Hoang, et al. 2020; Todd, et al. 2020; Palazzo, et al. 2022). You can clearly see in the example in Figure 6D the nucleus extending way beyond Otx2 expression as it is probably overlapping in space. Other examples should be shown, however, considering less than 1% of cells were putatively Otx2+, the safer interpretation is that these cells are not differentiating into neurons. At least 99.5% are not.

    We have additional examples of Otx2+ Tdt+ Edu+ cells, which suggest that MG neurogenesis to Otx2+ cells does occur, despite the low efficiency. We will include these images in the revised manuscript.

    (16) Same as above Figure 6I is not convincingly co-labeled HuC/D is an RNA-binding protein and unfortunately is not always the clearest stain but this looks like background haze in the INL overlapping. Other amacrine markers could be tested, but again due to the very low numbers, I think no neurogenesis is occurring.

    We have additional examples of HuC/D+ Tdt+ Edu+ cells, which we will show in the revised manuscript.

    (17) In the text the authors are accidently referring to Figure 6 as Figure 7.

    We thank the reviewer for pointing out the mistake. We will correct the mistake in the revised manuscript.

    Figure 7

    (18) I like this figure and the concept that you can have additional MG proliferating without destroying the retina or compromising vision. This is reminiscent of the chick MG reprogramming studies in which MG proliferate in large numbers and often do not differentiate into neurons yet still persist de-laminated for long time points.

    General:

    (19) The title should be changed, as I don't believe there is any convincing evidence of regeneration of neurons. Understanding the barriers to MG cell-cycle re-entry are important and I believe the authors did a good job in that respect, however it is an oversell to report regeneration of neurons from this data.

    We thank the reviewer for the suggestion. We will consider changing the title in the revised manuscript.

    (20) This paper uses multiple mouse lines and it is often confusing when the text and figures switch between models. I think it would be helpful to readers if the mouse strain was added to graphical paradigms in each figure when a different mouse line is employed.

    We will label the mouse lines used in each experiment in the figures where appropriate.