Synapse weakening-induced caspase-3 activity confers specificity to microglia-mediated synapse elimination

  1. Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, public reviews, and a provisional response from the authors.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Margaret Ho
    National Yang Ming Chiao Tung University, Taipei, Taiwan
  • Senior Editor
    Sofia Araújo
    University of Barcelona, Barcelona, Spain

Reviewer #1 (Public Review):

In this manuscript, the authors study the effects of synaptic activity on the process of eye-specific segregation, focusing on the role of caspase 3, classically associated with apoptosis. The method for synaptic silencing is elegant and requires intrauterine injection of a tetanus toxin light chain into the eye. The authors report that this silencing leads to increased caspase 3 in the contralateral eye (Figure 1) and demonstrate evidence of punctate caspase 3 that does not overlap neuronal markers like map2. However, the quantifications showing increased caspase 3 in the silenced eye (done at P5) are complicated by overlap with the signal from entire dying cells in the thalamus. The authors also show that global caspase 3 deficiency impairs the process of eye-specific segregation and circuit refinement (Figures 3-4).

The authors also report that "synapse weakening-induced caspase-3 activation determines the specificity of synapse elimination mediated by microglia but not astrocytes" (abstract). They report that microglia engulf fewer RGC axon terminals in caspase 3 deficient animals (Figure 5), and that this preferentially occurs in silenced terminals, but this preferential effect is lost in caspase 3 knockouts. Based on this, the authors conclude that caspase 3 directs microglia to eliminate weaker synapses. However, a much simpler and critical experiment that the authors did not perform is to eliminate microglia and show that the caspase 3 dependent effects go away. Without this experiment, there is no reason to assume that microglia are directing synaptic elimination.

Finally, the authors also report that caspase 3 deficiency alters synapse loss in 6-month-old female APP/PS1 mice, but this is not really related to the rest of the paper.

Reviewer #2 (Public Review):

Summary:

This manuscript by Yu et al. demonstrates that activation of caspase-3 is essential for synapse elimination by microglia, but not by astrocytes. This study also reveals that caspase 3 activation-mediated synapse elimination is required for retinogeniculate circuit refinement and eye-specific territories segregation in dLGN in an activity-dependent manner. Inhibition of synaptic activity increases caspase-3 activation and microglial phagocytosis, while caspase-3 deficiency blocks microglia-mediated synapse elimination and circuit refinement in the dLGN. The authors further demonstrate that caspase-3 activation mediates synapse loss in AD, loss of caspase-3 prevented synapse loss in AD mice. Overall, this study reveals that caspase-3 activation is an important mechanism underlying the selectivity of microglia-mediated synapse elimination during brain development and in neurodegenerative diseases.

Strengths:

A previous study (Gyorffy B. et al., PNSA 2018) has shown that caspase-3 signal correlates with C1q tagging of synapses (mostly using in vitro approaches), which suggests that caspase-3 would be an underlying mechanism of microglial selection of synapses for removal. The current study provides direct in vivo evidence demonstrating that caspase-3 activation is essential for microglial elimination of synapses in both brain development and neurodegeneration.

The paper is well-organized and easy to read. The schematic drawings are helpful for understanding the experimental designs and purposes.

Weaknesses:

It seems that astrocytes contain large amounts of engulfed materials from ipsilateral and contralateral axon terminals (Figure S11B) and that caspase-3 deficiency also decreased the volume of engulfed materials by astrocytes (Figures S11C, D). So the possibility that astrocyte-mediated synapse elimination contributes to circuit refinement in dLGN cannot be excluded.

Does blocking single or dual inactivation of synapse activity (using TeTxLC) increase microglial or astrocytic engulfment of synaptic materials (of one or both sides) in dLGN?

Author response:

The following is the authors’ response to the original reviews.

eLife Assessment

This study presents an important finding on the involvement of a Caspase 3-dependent pathway in the elimination of synapses for retinogeniculate circuit refinement and eye-specific territory segregation. This work fits well with the concept of "synaptosis" which has been proposed in the past but lacked in vivo support. Despite its elegant design and many strengths, the evidence supporting the claims of the authors is incomplete, particularly regarding whether Caspase-3 expression can really be isolated to synapses vs locally dying cells, whether microglia direct or instruct synapse elimination, and whether astrocytes are also involved. The work will be of interest to investigators studying cell death pathways, neurodevelopment, and neurodegenerative disease.

Regarding significance:

This study provides in vivo evidence that caspase-3 is important for synapse elimination in the visual pathway (Figure 3 and 4) and corroborates the previously proposed but not yet validated “synaptosis” hypothesis. But more significantly, we show that caspase-3 is activated in dLGN relay neurons in response to synapse inactivation (Figure 1) when synaptic competition is present (Figure 2), and that caspase-3 is important for efficient elimination of weakened synapses by microglia (Figure 5 and 6). We consider the causal link between synapse weakening/inactivation and caspase-3 activation to be the most important finding of this study and believe it is an error to not include this aspect of the study in the assessment. The mechanism by which neuronal activity influences synapse elimination is a fundamental question in neuroscience, and our study presents a significant advancement in understanding this problem.

Regarding strength of evidence:

We do not agree with the assessment that our evidence should be broadly labeled as “incomplete”. In fact, we argue that many concerns raised by the reviewers are not focused on the main claims made in this study.

(1) Regarding whether caspase-3 activation (not “expression”, which is the term used in the assessment) is isolated to synapses or occurs in entire cells, we show in Figure 1 that both types of signals can be present. The main concern of the reviewers seems to be that activated caspase-3 signals in apoptotic dLGN relay neurons are irrelevant to our analysis and confound interpretation. We argue that this is not the case.

In Figure 1, we have two sets of controls demonstrating that the observed apoptosis of dLGN relay neurons occurs specifically in response to synapse inactivation. For each animal that received TeTxLC injection in the right eye, activated caspase-3 signal is compared between the left dLGN, where most of the inactivated synapses are located, and the right dLGN, where the minority of the inactivated synapses are located (between Figure 1B and 1C, also between the first and second group of Figure 1E). We observed apoptotic neurons in the right dLGN with more inactivated synapses but not in the left dLGN with fewer inactivated synapses. The second control is between TeTxLC-injected animals (Figure 1B) and mock-injected animals (Figure 1D). We observed apoptotic relay neurons in the dLGN of TeTxLC-injected animals (Figure 1B) but not mock-injected animals (Figure 1D). Both these controls show that the observed apoptosis of dLGN relay neurons is caused by synapse inactivation.

In addition, in our synapse inactivation experiment (Figure 1), AAV-hSyn-TeTxLC is injected into the right eye and expressed only in RGCs, not in dLGN relay neurons. Since dLGN relay neurons in this experiment do not receive a perturbation that is independent of synaptic transmission, we conclude that their apoptosis occurs through synapse-dependent mechanisms.

Furthermore, if the apoptotic neurons are confounding the analysis (as implied by reviewers and editors) and do not occur through synapse-dependent mechanisms, then inhibiting both eyes with TeTxLC (Figure 2C, rightmost group) should cause high levels of caspase-3 activation, like that in the single-inhibition condition. Instead, we observe the opposite (Figure 2C, middle group) – overall caspase-3 activity goes down significantly in the dual-inhibition condition and is closer to the unperturbed condition, which can be explained by a loss of interaction between “strong” and “weak” synapses. Taken together, our data demonstrate that apoptosis of relay neurons in Figure 1 occurs specifically in response to synapse inactivation through synapse-dependent mechanisms, and the activated caspase-3 signal in the neurons should be included in our analysis.

Why does synaptic caspase-3 activation manifest in different forms: puncta, “blobs”, and cells? This is not surprising when considering the mechanisms that neurons must utilize to spatially confine caspase-3 activation and the nature of the apoptotic signaling cascade. On one hand, it has been proposed that caspase-3 activity in dendrites can be locally confined by proteasomal degradation of cleaved caspase-3 (Erturk et al., DOI: 10.1523/JNEUROSCI.3121-13.2014 ). On the other hand, caspase-3 activation is known to trigger explosive feedback amplification of apoptotic signaling events (McComb et al., DOI: 10.1126/sciadv.aau9433 ). For caspase-3 activation to remain localized to dendrites, the negative regulation must outweigh the positive feedback amplification. By expressing TeTxLC in RGCs of one eye, we create a strong perturbation that silences a large fraction of the synapses in the retinogeniculate pathway, which likely shifts the balance between positive and negative regulation of caspase-3 activity in some relay neurons. To be more specific, if a given dLGN relay neuron receives too many inactivated synapses, which is likely the case in our perturbation, caspase-3 activity that is initially localized can overwhelm the physiological negative regulation mechanisms that act to spatially confine it, resulting in whole cell apoptosis. In fact, previous in vitro evidence (Enturk et al., DOI: 10.1523/JNEUROSCI.3121-13.2014 ) demonstrated that, while caspase-3 activation in a single distal dendrite can be locally contained, activating apoptosis signaling in dendrites proximal to the cell body can result in whole-cell apoptosis. Similarly, a few inactivated retinogeniculate synapses can elicit locally contained caspase-3 activity in dLGN relay neurons, but a large number of inactivated synapses on a single relay neuron may trigger sufficient caspase-3 activity that can lead to whole-cell apoptosis. We discussed how to interpret synapse inactivation-induced apoptosis in dLGN relay neurons both in the main text and in the discussion (line 123-132, and line 411-421).

(2) Regarding microglia, we did not claim that “microglia direct or instruct synapse elimination”. Our main claim is that caspase-3 activation is important for efficient elimination of weakened synapses by microglia. This claim emphasizes a regulatory role for caspase-3 activation in microglia-mediated synapse elimination, but not a regulatory role of microglia in synapse elimination. To be more specific, our data suggest that lack of synaptic activity induces caspase-3 activity, and caspase-3 activity in turn influences which synapses are preferentially eliminated by microglia. Therefore, the elimination specificity is fundamentally determined (i.e. instructed) by neuronal activity, not by microglia. We also did not presume the manner in which microglia engage in synapse elimination. We specifically address this point in the discussion at line 458 through 465 where we acknowledge that microglia may indirectly mediate synapse elimination by engulfing shed neuronal material. In our title and text, we use the phrase “microglia-mediated synapse elimination”, which is not the same as microglia-instructed synapse elimination and does not presume any instructive/directive role of microglia.

(3) Regarding whether astrocytes are involved, we did not challenge the notion that astrocytes play important roles in synapse elimination. Rather, our claim is that, unlike what we observed with microglia, the amount of synaptic material engulfed by astrocytes does not robustly depend on whether caspase-3 is present. We acknowledge that there might be a caspase-3 dependent phenotype that we were unable to detect (line 309-310), and that it is plausible that astrocytes mediate activity-dependent synapse elimination through other caspase-3-independent mechanisms. This claim is not central to our study, and we would like to qualify the statements in the manuscript. We will remove the phrase “but not astrocytes” in line 18 of the abstract.

In summary, using a state-of-the-art method to inactivate retinogeniculate synapses, we discovered a causal link between synapse weakening/inactivation and caspase-3 activation. Coupled with well-established in vivo assays (e.g., segregation analysis, electrophysiology, and engulfment analysis) that are used in many landmark studies we cite, we provide solid evidence supporting our claim that “caspase-3 is essential for synapse elimination driven by both spontaneous and experience-dependent neural activity”, and that “synapse weakening-induced caspase-3 activation determines the specificity of synapse elimination mediated by microglia”.

Public Reviews:

Reviewer #1 (Public Review):

In this manuscript, the authors study the effects of synaptic activity on the process of eye-specific segregation, focusing on the role of caspase 3, classically associated with apoptosis. The method for synaptic silencing is elegant and requires intrauterine injection of a tetanus toxin light chain into the eye. The authors report that this silencing leads to increased caspase 3 in the contralateral eye (Figure 1) and demonstrate evidence of punctate caspase 3 that does not overlap neuronal markers like map2. However, the quantifications showing increased caspase 3 in the silenced eye (done at P5) are complicated by overlap with the signal from entire dying cells in the thalamus. The authors also show that global caspase 3 deficiency impairs the process of eye-specific segregation and circuit refinement (Figures 3-4).

The reviewer states: “this silencing leads to increased caspase 3 in the contralateral eye”. We observed increased caspase-3 activity, not protein levels, in the contralateral dLGN, not eye.

The reviewer states: “and demonstrate evidence of punctate caspase 3 that does not overlap neuronal markers like map2”. This is not accurate. We show that the punctate active caspase-3 signals overlap with the dendritic marker MAP2 (Figure S4A).

The reviewer states: “, the quantifications showing increased caspase 3 [activity] in the silenced [dLGN] (done at P5) are complicated by overlap with the signal from entire dying cells in the thalamus”. This is not accurate. The apoptotic neurons we observed are relay neurons located in the dLGN (confirmed by their morphology and positive staining of NeuN – Figure S4B-C), not “cells” of unknown lineage (as suggested by the reviewer) in the general “thalamus” area (as suggested by the reviewer). If the dying cells were non-neuronal cells, that would indeed confound our quantification and conclusions, but that is not the case.

We argue that the active caspase-3 signals in apoptotic dLGN relay neurons are not a confounding factor but a bona fide response to synaptic silencing and therefore should be included in the quantification. We have two sets of controls (please also see the general response above), one is between the strongly inactivated dLGN and the weakly inactivated dLGN in each TeTxLC-injected animal, second is between dLGN of TeTxLC-injected animals and mock-injected animals. In both controls, only the dLGN receiving strong synapse inactivation has these apoptotic dLGN relay neurons, demonstrating that these cells occur as a consequence of synapse inactivation. It is also unlikely that our perturbation is causing cell death through a non-synaptic mechanism. As mock injections do not cause apoptosis in dLGN neurons, this phenomenon is not related to surgical damage. TeTxLC is injected into the eyes and only expressed in presynaptic RGCs, not in postsynaptic relay neurons, so this phenomenon is also unlikely to be caused by TeTxLC-related toxicity. Furthermore, if apoptosis of dLGN relay neurons is not related to synapse inactivation, then when TeTxLC is injected into both eyes, one would expect to see either the same amount or more apoptotic relay neurons, but we instead observed a reduction in dLGN neuron apoptosis, suggesting a synapse-related mechanism must be responsible. Considering the above, apoptosis of relay neurons in TeTxLC-inactivated dLGN is causally linked to synapse inactivation, and active caspase-3 signals in these neurons are true signals that should be included in the quantification.

The authors also report that "synapse weakening-induced caspase-3 activation determines the specificity of synapse elimination mediated by microglia but not astrocytes" (abstract). They report that microglia engulf fewer RGC axon terminals in caspase 3 deficient animals (Figure 5), and that this preferentially occurs in silenced terminals, but this preferential effect is lost in caspase 3 knockouts. Based on this, the authors conclude that caspase 3 directs microglia to eliminate weaker synapses. However, a much simpler and critical experiment that the authors did not perform is to eliminate microglia and show that the caspase 3 dependent effects go away. Without this experiment, there is no reason to assume that microglia are directing synaptic elimination.

The reviewer states: “microglia engulf fewer RGC axon terminals in caspase 3 deficient animals (Figure 5), and that this preferentially occurs in silenced terminals, but this preferential effect is lost in caspase 3 knockouts”. We are not sure what the reviewer means by “this preferentially occurs in silenced terminals”. Our results show that microglia preferentially engulf silenced terminals, and such preference is lost in caspase-3 deficient mice (Figure 6).

We do not understand the experiment where the reviewer suggested to: “eliminate microglia and show that the caspase 3 dependent effects go away”. To quantify caspase-3 dependent engulfment of synaptic material by microglia or preferential engulfment of silenced terminals by microglia, microglia must be present in the tissue sample. If we eliminate microglia, neither of these measurements can be made. What could be measured if microglia are eliminated is the refinement of retinogeniculate pathway. This experiment would test whether microglia are required for caspase-3 dependent phenotypes. This is not a claim made in the manuscript. Instead, we claimed caspase-3 is required for microglia to preferentially eliminate weak synapses.

We did not claim that “microglia are directing synaptic elimination”. Our claim is that synapse inactivation induces caspase-3 activity, and this caspase-3 activity in turn determines the substrate preference of microglia-mediated synapse elimination. Based on this model, it is the neuronal activity that fundamentally directs synapse elimination. Throughout the manuscript, we used the term “microglia-mediated synapse elimination”. This terminology does not assume a directive/instructive role of microglia in synapse elimination and only describes the observed engulfment of synaptic material by microglia. We also did not assume how microglia engage in synapse elimination. We acknowledge in the discussion (line 458 through 465) that microglia may mediate synapse elimination in an indirect, passive way by engulfing shed neuronal material. This topic is a matter of debate in the field (Eyo et al., DOI: 10.1126/science.adh7906 ).

Finally, the authors also report that caspase 3 deficiency alters synapse loss in 6-month-old female APP/PS1 mice, but this is not really related to the rest of the paper.

We respectfully disagree that Figure 7 is not related to the rest of the paper. Many genes involved in postnatal synapse elimination, such as C1q and C3, have been implicated in neurodegeneration. It is therefore natural and important to ask whether the function of caspase-3 in regulating synaptic homeostasis extends to neurodegenerative diseases in adult animals. The answer to this question may have broad therapeutic impacts.

Reviewer #2 (Public Review):

Summary:

This manuscript by Yu et al. demonstrates that activation of caspase-3 is essential for synapse elimination by microglia, but not by astrocytes. This study also reveals that caspase 3 activation-mediated synapse elimination is required for retinogeniculate circuit refinement and eye-specific territories segregation in dLGN in an activity-dependent manner. Inhibition of synaptic activity increases caspase-3 activation and microglial phagocytosis, while caspase-3 deficiency blocks microglia-mediated synapse elimination and circuit refinement in the dLGN. The authors further demonstrate that caspase-3 activation mediates synapse loss in AD, loss of caspase-3 prevented synapse loss in AD mice. Overall, this study reveals that caspase-3 activation is an important mechanism underlying the selectivity of microglia-mediated synapse elimination during brain development and in neurodegenerative diseases.

Strengths:

A previous study (Gyorffy B. et al., PNSA 2018) has shown that caspase-3 signal correlates with C1q tagging of synapses (mostly using in vitro approaches), which suggests that caspase-3 would be an underlying mechanism of microglial selection of synapses for removal. The current study provides direct in vivo evidence demonstrating that caspase-3 activation is essential for microglial elimination of synapses in both brain development and neurodegeneration.

The paper is well-organized and easy to read. The schematic drawings are helpful for understanding the experimental designs and purposes.

Weaknesses:

It seems that astrocytes contain large amounts of engulfed materials from ipsilateral and contralateral axon terminals (Figure S11B) and that caspase-3 deficiency also decreased the volume of engulfed materials by astrocytes (Figures S11C, D). So the possibility that astrocyte-mediated synapse elimination contributes to circuit refinement in dLGN cannot be excluded.

The experiments presented in Figure S11 aim to determine whether astrocyte-mediated synapse elimination depends on caspase- 3 signaling. We do not claim that astrocytes are unimportant for synapse elimination or circuit refinement. We did observe a small decrease in synaptic material engulfed by astrocytes when caspase-3 is deficient, and we acknowledged that there could be defects that we were not able to detect (line 309-310). The claim that caspase-3 does not regulate astrocyte-mediated synapse elimination is not a central claim of the manuscript and we will qualify our statements in the text. We will remove the phrase “but not astrocytes” in the abstract (line 18).

Does blocking single or dual inactivation of synapse activity (using TeTxLC) increase microglial or astrocytic engulfment of synaptic materials (of one or both sides) in dLGN?

We assume that by “blocking single or dual inactivation of synapse activity”, the reviewer refers to inactivating retinogeniculate synapses from one or both eyes.

We showed that inactivating retinogeniculate synapses from one eye (single inactivation) increases microglia-mediated engulfment of presynaptic terminals of inactivated synapses (Figure 6). We did not measure microglia-mediated engulfment of synaptic material while inactivating retinogeniculate synapses from both eyes (dual inactivation). However, based on the total active caspase-3 signal (Figure 2) in the dual inactivation scenario, we do not expect to see an increase in engulfment of synaptic material.

We did not measure astrocyte-mediated engulfment with single or dual inactivation, as we did not see a robust caspase-3 dependent phenotype in astrocyte-mediated engulfment.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation