GluA3 subunits are required for appropriate assembly of AMPAR GluA2 and GluA4 subunits on cochlear afferent synapses and for presynaptic ribbon modiolar–pillar morphology

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife assessment

    Hearing is mediated by hair cells in the cochlea, which synapse onto the primary dendrites of the auditory nerve. This study shows how deletion of a postsynaptic glutamate receptor subtype strongly influences inner hair cell-spiral ganglion cell synapse formation. Thus pre- and post-synaptic changes are dynamically intertwined, providing insights into how pathological outcomes arise from synaptic perturbations.

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

Cochlear sound encoding depends on α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs), but reliance on specific pore-forming subunits is unknown. With 5-week-old male C57BL/6J Gria3 -knockout mice (i.e., subunit GluA3 KO ) we determined cochlear function, synapse ultrastructure, and AMPAR molecular anatomy at ribbon synapses between inner hair cells (IHCs) and spiral ganglion neurons. GluA3 KO and wild-type (GluA3 WT ) mice reared in ambient sound pressure level (SPL) of 55–75 dB had similar auditory brainstem response (ABR) thresholds, wave-1 amplitudes, and latencies. Postsynaptic densities (PSDs), presynaptic ribbons, and synaptic vesicle sizes were all larger on the modiolar side of the IHCs from GluA3 WT , but not GluA3 KO , demonstrating GluA3 is required for modiolar–pillar synapse differentiation. Presynaptic ribbons juxtaposed with postsynaptic GluA2/4 subunits were similar in quantity, however, lone ribbons were more frequent in GluA3 KO and GluA2-lacking synapses were observed only in GluA3 KO . GluA2 and GluA4 immunofluorescence volumes were smaller on the pillar side than the modiolar side in GluA3 KO , despite increased pillar-side PSD size. Overall, the fluorescent puncta volumes of GluA2 and GluA4 were smaller in GluA3 KO than GluA3 WT . However, GluA3 KO contained less GluA2 and greater GluA4 immunofluorescence intensity relative to GluA3 WT (threefold greater mean GluA4:GluA2 ratio). Thus, GluA3 is essential in development, as germline disruption of Gria3 caused anatomical synapse pathology before cochlear output became symptomatic by ABR. We propose the hearing loss in older male GluA3 KO mice results from progressive synaptopathy evident in 5-week-old mice as decreased abundance of GluA2 subunits and an increase in GluA2-lacking, GluA4-monomeric Ca 2+ -permeable AMPARs.

Article activity feed

  1. Author Response

    Reviewer #1 (Public Review):

    It has previously been shown that deletion of the GluA3 subunit in mice leads to alterations in auditory behavior in adult mice that are older than a couple of months of age. The GluA3 subunit is expressed at several synapses along the auditory pathway (cochlea and brainstem), and in ko mice changes in brainstem synapses have been observed. These previously documented changes may account for some of the deficits in hearing in adult ko mice.

    In the current study, the authors investigate an earlier stage of development (at 5 wks) when the auditory brainstem responses (ABRs) are normal, and they ask how transmission persists at inner hair cell (ihc) ribbon synapses in GluA3 ko mice. They discovered that deletion of GluR3A significantly changed 1) the relative expression of Glu A2 (dramatically downregulated) and A4 subunits at SGN afferents, and 2) caused morphological changes in ihc ribbons (modiolar side) and synaptic vesicle size (pillar).

    The changes documented in the 5 wk old GluA3ko mice were not necessarily predicted because in general the mechanisms involved in shuffling GluA receptors at this synapse (or other sensory synapses) are not completely understood; furthermore, much less is known about the role of differentiation of ihc-sgn synapses along a modiolar-pillar axis. With that said, the only shortcoming of the study is a lack of explanation for the observed changes in the synaptic structure; but this is not specific to this study.

    Given the quality of the data and the clarity of presentation of results, this is a very valuable study that will aid and motivate researchers to further explore how auditory circuitry develops, and becomes differentiated, at the level of ihc-sgn synapses.

    We thank the reviewer for the positive and helpful comments. Ongoing studies are seeking to explain the observed changes in synapse structure.

    Reviewer #2 (Public Review):

    The goal of the study by Rutherford and colleagues was to characterize functional, structural, and molecular changes at the highly specialized cochlear inner hair cell (IHC) - spiral ganglion neuron (SGN) ribbon synapse in GluA3 AMPA receptor subunit knockout mice (GluA3KO). Previous work by the authors demonstrated that 2-month-old GluA3KO mice experienced impaired auditory processing and changes in synaptic ultrastructure at the SGN - bushy cell synapse, the next synapse in the auditory pathway.

    In the present study, the authors investigated whether GluA3 is required for ribbon synapse formation and physiology in 5-week-old mice using a series of functional and light- and electron microscopy imaging approaches. While deletion of GluA3 AMPAR subunit did not affect hearing sensitivity at this age, the authors reported that cochlear ribbon synapses exhibited changes in the molecular composition of AMPARs and pre- and postsynaptic ultrastructural alterations. Specifically, the authors demonstrated that GluA3KO ribbon synapses exhibit i) a global reduction in postsynaptic AMPARs, which is also reflected by smaller AMPAR arrays, ii) a reduction in GluA2 and an increase in GluA4 protein expression at individual postsynaptic sites, and iii) changes in the dimensions and morphology of the presynaptic specialization ("ribbon") and in the size of synaptic vesicles. These reported structural changes are linked to the side of innervation with respect to the IHC modiolar-pillar axis.

    The results presented by the authors are conceptually very interesting as the data support the notion that potentially detrimental changes in the molecular composition of a sensory synapse can be compensated to sustain synaptic function to a certain extent during development. The conclusions of the study are mostly well supported by the data, but some experimental details or control experiments are missing or need to be clarified to allow a full assessment.

    1. The authors tested which GluA isoforms are expressed in SGNs of GluA3KO mice and reported that only GluA2 and GluA4, and not GluA1, receptor subunits are present in the cochlear. It is, however, a bit difficult to understand why immunolabelling for GluA1 was only performed on brainstem sections (Fig. 1B right) and not in the cochlear to probe for postsynaptic localization at ribbon synapses as it was done for the other isoforms (Fig. 2 and 6) given that GluA3KO IHCs exhibited a larger number of ribbons that lacked GluA2 and 3 (lone or 'orphaned' ribbons; Fig. 6B). It is also not clear why immunolabelling for GluA2 and 4 was performed to probe for expression of these receptor subunits on SGN cell bodies in the cochlear spiral ganglion. Which neurons are expected to synapse onto these somata?

    There is precedent for expression of GluA subunits in the SGN cell bodies reflecting expression at the synapse, although it is not clear if any of that immunoreactivity reflects cell surface expression in the intact ganglion or if it represents solely intracellular subunits being trafficked to synapses.

    Figure 1b shows that GluA2 is expressed in the somata of WT mice and KO mice. The lower panels show that GluA1 is not expressed in the somata of WT or KO mice. The right panels show that while GluA1 is expressed in the cerebellum of WT and KO mice, is not expressed in the cochlear nucleus of WT or KO mice. We think this demonstrates the lack of compensation by GluA1 in the GluA3 KO.

    We have now added GluA4 immunoreactivity in the SGNs to Fig. 1, for completeness. In our experience, GluA subunits expressed at synapses are also found in the cell bodies, and GluA subunits not expressed at synapses are not found in the cell bodies. The current data is consistent with this, although we did not label GluA1 in the organ of Corti.

    1. The authors state in the text that GluA3 expression is completely abolished in GluA3KO IHCs, however, there appears to still be a faint punctate immunofluorescence signal visible when an antibody directed against GluA3 was used (Fig. 2C). Providing additional information on the specificity of this (and the other) antibodies used in the study would be helpful.

    We agree, and thank the reviewer for pointing this out. There is indeed a small signal presumably due to cross-reactivity of the anti-GluA3 with GluA2 subunits, because the cytoplasmic epitope recognized by the antibody is in a region of high similarity of GluA2 and GluA3 (Dong et al., 1997). In addition, the specification sheet of the Santa Cruz company states that the GluA3 antibody can detect GluA2. This relatively small cross-reactivity is noted now in the text on p. 9. Also, this appearance was a product of the same brightness and contrast issue noted above in the response to the editor’s summary. Upon readjustment, the signal is less apparent, because in the readjustment we used less brightness and less contrast enhancement to avoid the unwanted saturation in some of the panels.

    1. The authors reported changes in the volume of the presynaptic ribbon and postsynaptic density surface area in GluA3KO KO animals. The EM data as presented are however not sufficiently convincing.

    i) There appears to be a mismatch between the EM data shown in Fig. 3 and 4 and the information in the text with respect to the number of data points in the plots and the reported number of reconstructed synapses. This raises several questions with respect to the analysis. For instance, it is unclear whether certain synapses were reconstructed but excluded from the analysis. If so, what were the exclusion criteria?

    We thank the reviewer for pointing out this discrepancy within the text and the figures. The discrepancies are now fixed. We have added more information on how the synapses were reconstructed in the M&M (p.14-15).

    ii) The authors compare PSD surface areas in reconstructions from 3D serial sections, but for some of the shown reconstructions (i.e. Fig. 3A' and B' and 4B'), it appears as if PSDs were only incompletely reconstructed.

    We included all the ultrathin sections that show afferent dendrites with a visible PSD. We revised all the reconstructions and fixed some misalignments. The appearance of the reconstructed PSD relates to how the Reconstruct software creates the 3-D rendering. We did not use any extra software to smooth the hedges of the 3D reconstructions.

    1. The immunolabelling experiments shown in Fig. 2 and 6 are of very high quality and the quantitative analysis of the light microscopy data (Fig. 6-9) is clearly very detailed, but slightly difficult to interpret the way it is presented. Specifically, it is unclear how the number of synapses per IHC (Fig. 6B) and the separation into modiolar and pillar side (Fig. 8) was achieved based on the shown images without the outlines of individual cells being visible.

    We agree. Please see the revised Figs. 2, 6, and 8, and explanation in the figure legend of Fig. 8.

    1. Adding more detailed information about important parameters (mean, N/n, SD/SEM) and the statistical tests used for the individual comparisons presented in the Figures would help strengthen the confidence in the presented data.

    Please see the new spreadsheets accompanying the revised manuscript.

    1. In general, the authors report a series of molecular and structural changes in IHCs and reach the conclusion that GluA3 subunits may have a role in "trans-synaptically" determining or organizing the architecture of both the pre- and post-synapse. However, some of the arguments are very speculative and many of the claims are not supported by experimental data presented in the paper. The authors should consider to also compare their findings to studies that investigated ultrastructural changes of AMPAR subunit knockouts in other synapse types, and discuss alternative interpretations (e.g. homeostatic changes).

    Thank you for this comment. Considering that reviewer 1 asked for more speculation, we have decided to leave the level of speculation similar to the initial submission. However, we went through the text to make sure our claims were backed by our observations.

    Due to space constraints, rather than comparing to additional other synapses, in this context we prefer to compare with auditory brainstem synapses.

    The possibility of homeostatic changes we now added on p. 29.

  2. eLife assessment

    Hearing is mediated by hair cells in the cochlea, which synapse onto the primary dendrites of the auditory nerve. This study shows how deletion of a postsynaptic glutamate receptor subtype strongly influences inner hair cell-spiral ganglion cell synapse formation. Thus pre- and post-synaptic changes are dynamically intertwined, providing insights into how pathological outcomes arise from synaptic perturbations.

  3. Reviewer #1 (Public Review):

    It has previously been shown that deletion of the GluA3 subunit in mice leads to alterations in auditory behavior in adult mice that are older than a couple of months of age. The GluA3 subunit is expressed at several synapses along the auditory pathway (cochlea and brainstem), and in ko mice changes in brainstem synapses have been observed. These previously documented changes may account for some of the deficits in hearing in adult ko mice.

    In the current study, the authors investigate an earlier stage of development (at 5 wks) when the auditory brainstem responses (ABRs) are normal, and they ask how transmission persists at inner hair cell (ihc) ribbon synapses in GluA3 ko mice. They discovered that deletion of GluR3A significantly changed 1) the relative expression of Glu A2 (dramatically downregulated) and A4 subunits at SGN afferents, and 2) caused morphological changes in ihc ribbons (modiolar side) and synaptic vesicle size (pillar).

    The changes documented in the 5 wk old GluA3ko mice were not necessarily predicted because in general the mechanisms involved in shuffling GluA receptors at this synapse (or other sensory synapses) are not completely understood; furthermore, much less is known about the role of differentiation of ihc-sgn synapses along a modiolar-pillar axis. With that said, the only shortcoming of the study is a lack of explanation for the observed changes in the synaptic structure; but this is not specific to this study.

    Given the quality of the data and the clarity of presentation of results, this is a very valuable study that will aid and motivate researchers to further explore how auditory circuitry develops, and becomes differentiated, at the level of ihc-sgn synapses.

  4. Reviewer #2 (Public Review):

    The goal of the study by Rutherford and colleagues was to characterize functional, structural, and molecular changes at the highly specialized cochlear inner hair cell (IHC) - spiral ganglion neuron (SGN) ribbon synapse in GluA3 AMPA receptor subunit knockout mice (GluA3KO). Previous work by the authors demonstrated that 2-month-old GluA3KO mice experienced impaired auditory processing and changes in synaptic ultrastructure at the SGN - bushy cell synapse, the next synapse in the auditory pathway.

    In the present study, the authors investigated whether GluA3 is required for ribbon synapse formation and physiology in 5-week-old mice using a series of functional and light- and electron microscopy imaging approaches. While deletion of GluA3 AMPAR subunit did not affect hearing sensitivity at this age, the authors reported that cochlear ribbon synapses exhibited changes in the molecular composition of AMPARs and pre- and postsynaptic ultrastructural alterations. Specifically, the authors demonstrated that GluA3KO ribbon synapses exhibit i) a global reduction in postsynaptic AMPARs, which is also reflected by smaller AMPAR arrays, ii) a reduction in GluA2 and an increase in GluA4 protein expression at individual postsynaptic sites, and iii) changes in the dimensions and morphology of the presynaptic specialization ("ribbon") and in the size of synaptic vesicles. These reported structural changes are linked to the side of innervation with respect to the IHC modiolar-pillar axis.

    The results presented by the authors are conceptually very interesting as the data support the notion that potentially detrimental changes in the molecular composition of a sensory synapse can be compensated to sustain synaptic function to a certain extent during development. The conclusions of the study are mostly well supported by the data, but some experimental details or control experiments are missing or need to be clarified to allow a full assessment.

    1. The authors tested which GluA isoforms are expressed in SGNs of GluA3KO mice and reported that only GluA2 and GluA4, and not GluA1, receptor subunits are present in the cochlear. It is, however, a bit difficult to understand why immunolabelling for GluA1 was only performed on brainstem sections (Fig. 1B right) and not in the cochlear to probe for postsynaptic localization at ribbon synapses as it was done for the other isoforms (Fig. 2 and 6) given that GluA3KO IHCs exhibited a larger number of ribbons that lacked GluA2 and 3 (lone or 'orphaned' ribbons; Fig. 6B). It is also not clear why immunolabelling for GluA2 and 4 was performed to probe for expression of these receptor subunits on SGN cell bodies in the cochlear spiral ganglion. Which neurons are expected to synapse onto these somata?

    2. The authors state in the text that GluA3 expression is completely abolished in GluA3KO IHCs, however, there appears to still be a faint punctate immunofluorescence signal visible when an antibody directed against GluA3 was used (Fig. 2C). Providing additional information on the specificity of this (and the other) antibodies used in the study would be helpful.

    3. The authors reported changes in the volume of the presynaptic ribbon and postsynaptic density surface area in GluA3KO KO animals. The EM data as presented are however not sufficiently convincing.
    i) There appears to be a mismatch between the EM data shown in Fig. 3 and 4 and the information in the text with respect to the number of data points in the plots and the reported number of reconstructed synapses. This raises several questions with respect to the analysis. For instance, it is unclear whether certain synapses were reconstructed but excluded from the analysis. If so, what were the exclusion criteria?
    ii) The authors compare PSD surface areas in reconstructions from 3D serial sections, but for some of the shown reconstructions (i.e. Fig. 3A' and B' and 4B'), it appears as if PSDs were only incompletely reconstructed.

    4. The immunolabelling experiments shown in Fig. 2 and 6 are of very high quality and the quantitative analysis of the light microscopy data (Fig. 6-9) is clearly very detailed, but slightly difficult to interpret the way it is presented. Specifically, it is unclear how the number of synapses per IHC (Fig. 6B) and the separation into modiolar and pillar side (Fig. 8) was achieved based on the shown images without the outlines of individual cells being visible.

    5. Adding more detailed information about important parameters (mean, N/n, SD/SEM) and the statistical tests used for the individual comparisons presented in the Figures would help strengthen the confidence in the presented data.

    6. In general, the authors report a series of molecular and structural changes in IHCs and reach the conclusion that GluA3 subunits may have a role in "trans-synaptically" determining or organizing the architecture of both the pre- and post-synapse. However, some of the arguments are very speculative and many of the claims are not supported by experimental data presented in the paper. The authors should consider to also compare their findings to studies that investigated ultrastructural changes of AMPAR subunit knockouts in other synapse types, and discuss alternative interpretations (e.g. homeostatic changes).