Ezrin defines TSC1 activation at endosomal compartments through EGFR-AKT signaling

  1. Department of Biology, University of Naples Federico II, Strada Vicinale Cupa Cintia, 21, Naples 80126 Italy
  2. Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy
  3. U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147, Genova, Italy
  4. Clinical Medicine and Surgery, University of Naples Federico II, Via S. Pansini 5, 80131 Naples Italy
  5. Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, Genova, Italy

Peer review process

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

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Elizabeth Henske
    Brigham And Women's Hospital, Boston, United States of America
  • Senior Editor
    Jonathan Cooper
    Fred Hutchinson Cancer Research Center, Seattle, United States of America

Reviewer #1 (Public Review):

Summary:
The authors demonstrate that, while the loss of Ezrin increases lysosomal biogenesis and function, its presence is required for the specific endocytosis of EGFR. Upon further investigation, the authors reveal that Ezrin is a crucial intermediary protein that links EGFR to AKT, leading to the phosphorylation and inhibition of TSC. TSC is a critical negative regulator of the mTORC1 complex, which is dysregulated in various diseases, making their findings a valuable addition to multiple fields of study. Their cell signaling findings are translatable to an in vivo Medaka fish model and suggest that Ezrin may play a crucial role in retinal degeneration.

Strengths:
Giamundo, Intartaglia, et al. utilized unbiased proteomic and transcriptomic screens in Ezrin KO cells to investigate the mechanistic function of Ezrin in lysosome and cell signaling pathways. The authors' findings are consistent with past literature demonstrating Ezrin's role in the EGFR and mTORC1 signaling pathways. They used several cell lines, small molecule inhibitors, and cellular and in vivo knockout models to validate signaling changes through biochemical and microscopy assays. Their use of multiple advanced microscopy techniques is also impressive.

Weaknesses:
While the authors demonstrated activation of TSC1 (lysosomal accumulation) and inactivation of Akt (decreased phosphorylation in TSC1), as well as decreased mTORC1 signaling in Ezrin knockout cells, direct experiments showing the rescue of mTORC1 activity by AKT and TSC1 mutants are required to confirm the linear signaling pathway and establish Ezrin as a mediator of EGFR-AKT-TSC1-mTORC1 signaling. Although the authors presented representative images from advanced microscopy techniques to support their claims, there is insufficient quantification of these experiments. Additionally, several immunoblots in the manuscript lack vital loading controls, such as input lanes for immunoprecipitations and loading controls for western blots.

Reviewer #2 (Public Review):

Summary:
The authors begin with the stated goal of gaining insight into the known repression of autophagy by Ezrin, a major membrane-actin linker that assembles signaling complexes on membranes. RNA and protein expression analysis is consistent with upregulation of lysosomal proteins in Ezrin-deficient MEFs, which the authors confirm by immunostaining and western blotting for lysosomal markers. Expression analysis also implicates EGF signaling as being altered downstream of Ezrin loss, and the authors demonstrate that Ezrin promotes relocalization of EGFR from the plasma membrane to endosomes. Ezrin loss impacts downstream MAPK/Akt/mTORC1 signaling, although the mechanistic links remain unclear. An Ezrin mutant Medaka fish line wa then generated to test Ezrin's role in retinal cells, which are known to be sensitive to changes in autophagy regulation. Phenotypes in this model appear generally consistent with observations made in cultured cells, though mild overall.

Strengths:
Data on the impact of Ezrin-loss on relocalization of EGFR from the plasma membrane are extensive, and thoroughly demonstrate that Ezrin is required for EGFR internalization in response to EGF.

A new Ezrin-deficient in vivo model (Medaka fish) is generated.

Strong data demonstrates that Ezrin loss suppresses Akt signaling. Ezrin loss also clearly suppresses mTORC1 signaling in cell culture, although examination of mTORC1 activity is notably missing in Ezrin-deficient fish.

Weaknesses:
LC3 is used as a readout of autophagy, however the lipidated/unlipidated LC3 ratio generally does not appear to change, thus there does not appear to be evidence that Ezrin loss is affecting autophagy in this study.

The conclusion is drawn that Ezrin loss suppresses EGF signaling, however this is complicated by a strong increase in phosphorylation of the p38 MAPK substrate MK2. Without additional characterization of MAPK and Erk signaling, the effect of Ezrin loss remains unclear.

Causative conclusions between effects on MAPK, Akt, and mTORC1 signaling are frequently drawn, but the data only demonstrate correlations. For example, many signaling pathways can activate mTORC1 including MAPK/Erk, thus reduced mTORC1 activity upon Ezrin-loss cannot currently be attributed to reduced Akt signaling. Similarly, other kinases can phosphorylate TSC2 at the sites examined here, so the conclusion cannot be drawn that Ezrin-loss causes a reduction in Akt-mediated TSC2 phosphorylation. In Figure 7, the conclusion cannot be drawn that retinal degeneration results from aberrant EGFR signaling.

It is unclear why TSC1 is highlighted in the title, as there does not appear to be any specific regulation of TSC1 here.

In Figure 1 the conclusion is drawn that there is an increase in lysosome number with Ezrin KO, however it does not appear that the current analysis can distinguish an increased number from increased lysosome size or activity. Similarly, conclusions about increased lysosome "biogenesis" could instead reflect decreased turnover.

Immunoprecipitation data for a role for Ezrin as a signaling scaffold appear minimal and seem to lack important controls.

In Figure 3A it seems difficult to conclude that EGFR dimerization is reduced since the whole blot, including the background between lanes, is lighter on that side.

In Figure 6C specificity controls for the TSC1 and TSC2 antibodies are not included, but seem necessary since their localization patterns appear very different from each other in WT cells.

In Figure 7 the signaling effects in Ezrin-deficient fish are mild compared to cultured cells, and effects on mTORC1 are not examined. Further data on the retinal cell phenotypes would strengthen the conclusions.

In Figure 7F there appears to be more EGFR throughout the cell, so it is difficult to conclude that more EGFR at the PM in Ezrin-/- fish means reduced internalization.

Reviewer #3 (Public Review):

Summary:
In this study, the authors have attempted to demonstrate a critical role for the cytoskeletal scaffold protein Ezrin, in the upstream regulation of EGFR/AKT/MTOR signaling. They show that in the absence of Ezrin, ligand-induced EGFR trafficking and activation at the endosomes is perturbed, with decreased endosomal recruitment of the TSC complex, and a corresponding decrease in AKT/MTOR signaling.

Strengths:
The authors have used a combination of novel imaging techniques, as well as conventional proteomic and biochemical assays to substantiate their findings. The findings expand our understanding of the upstream regulators of the EGFR/AKT MTOR signaling and lysosomal biogenesis, appear to be conserved in multiple species, and may have important implications for the pathogenesis and treatment of diseases involving endo-lysosomal function, such as diabetes and cancer, as well as neuro-degenerative diseases like macular degeneration. Furthermore, pharmacological targeting of Ezrin could potentially be utilized in diseases with defective TFEB/TFE3 functions like LSDs. While a majority of the findings appear to support the hypotheses, there are substantial gaps in the findings that could be better addressed. Since Ezrin appears to directly regulate MTOR activity, the effects of Ezrin KO on MTOR-regulated, TFEB/TFE3 -driven lysosomal function should be explored more thoroughly. Similarly, a more convincing analysis of autophagic flux should be carried out. Additionally, many immunoblots lack key controls (Control IgG in co-IPs) and many others merit repetition to either improve upon the quality of the existing data, validate the findings using orthogonal approaches, or provide a more rigorous quantitative assessment of the findings, as highlighted in the recommendation for authors.

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