Rapid and Inducible Mislocalization of Endogenous TDP43 in a Novel Human Model of Amyotrophic Lateral Sclerosis

  1. Living Systems Institute, University of Exeter, EX4 4QD, UK
  2. Biosciences, University of Exeter, EX4 4QD, UK
  3. Clinical and Biomedical Sciences, University of Exeter, EX4 7BS, UK

Editors

  • Reviewing Editor
    Hugo Bellen
    Baylor College of Medicine, Houston, United States of America
  • Senior Editor
    Tony Yuen
    Icahn School of Medicine at Mount Sinai, New York, United States of America

Reviewer #1 (Public Review):

Summary: Nuclear depletion and cytoplasmic mislocalization/aggregation of the DNA and RNA binding protein TDP-43 are pathological hallmarks of multiple neurodegenerative diseases. Prior work has demonstrated that depletion of TDP-43 from the nucleus leads to alterations in transcription and splicing. Conversely, cytoplasmic mislocalization/aggregation can contribute to toxicity by impairing mRNA transport and translation as well as miRNA dysregulation. However, to date, models of TDP-43 proteinopathy rely on artificial knockdown- or overexpression-based systems to evaluate either nuclear loss or cytoplasmic gain of function events independently. Few model systems authentically reproduce both nuclear depletion and cytoplasmic miscloalization/aggreagtion events. In this manuscript, the authors generate novel iPSC-based reagents to manipulate the localization of endogenous TDP-43. This is a valuable resource for the field to study pathological consequences of TDP-43 proteinopathy in a more endogenous and authentic setting. However, in the current manuscript, there are a number of weaknesses that should be addressed to further validate the ability of this model to replicate human disease pathology and demonstrate utility for future studies.

Strengths: The primary strength of this paper is the development of a novel in vitro tool.

Weaknesses: There are a number of weaknesses detailed below that should be addressed to thoroughly validate these new reagents as more authentic models of TDP-43 proteinopathy and demonstrate their utility for future investigations.

(1) The authors should include images of their engineered TDP-43-GFP iPSC line to demonstrate TDP-43 localization without the addition of any nanobodies (perhaps immediately prior to addition of nanobodies). Additionally, it is unclear whether simply adding a GFP tag to endogenous TDP-43 impact its normal function (nuclear-cytoplasmic shuttling, regulation of transcription and splicing, mRNA transport etc).

(2) Can the authors explain why there is a significant discrepancy in time points selected for nanobody transduction and immunostaining or cell lysis throughout Figure 1 and 2? This makes interpretation and overall assessment of the model challenging.

(3) The authors should further characterize their TDP-43 puncta. TDP-43 immunostaining is typically punctate so it is unclear if the puncta observed are physiologic or pathologic based on the analyses carried out in the current version of this manuscript. Additionally, do these puncta co-localize with stress granule markers or RNA transport granule markers? Are these puncta phosphorylated (which may be more reminiscent of end-stage pathologic observations in humans)?

(4) The authors should include multiple time points in their evaluation of TDP-43 loss of function events and aggregation. Does loss of function get worse over time? Is there a time course by which RNA misprocessing events emerge or does everything happen all at once? Does aggregation get worse over time? Do these neurons die at any point as a result of TDP-43 proteinopathy?

(5) Can the authors please comment on whether or not their model is "tunable"? In real human disease, not every neuron displays complete nuclear depletion of TDP-43. Instead there is often a gradient of neurons with differing magnitudes of nuclear TDP-43 loss. Additionally, very few neurons (5-10%) harbor cytoplasmic TDP-43 aggregates at end-stage disease. These are all important considerations when developing a novel authentic and endogenous model of TDP-43 proteinopathy which the current manuscript fails to address.

Reviewer #2 (Public Review):

Summary:
TDP-43 mislocalization occurs in nearly all of ALS, roughly half of FTD, and as a co-pathology in roughly half of AD cases. Both gain-of-function and loss-of-function mechanisms associated with this mislocalization likely contribute to disease pathogeneisis.

Here, the authors describe a new method to induce TDP-43 mislocalization in cellular models. They endogenously-tagged TDP-43 with a C-terminal GFP tag in human iPSCs. They then expressed an intrabody - fused with a nuclear export signal (NES) - that targeted GFP to the cytosol. Expression of this intrabody-NES in human iPSC-derived neurons induced nuclear depletion of homozygous TDP-43-GFP, caused its mislocalization to the cytosol, and at least in some cells appeared to cause cytosolic aggregates. This mislocalization was accompanied by induction of cryptic exons in well characterized transcripts known to be regulated by TDP-43, a hallmark of functional TDP-43 loss and consistent with pathological nuclear TDP-43 depletion. Interestingly, in heterozygous TDP-43-GFP neurons, expression of intrabody-NES appeared to also induce the mislocalization of untagged TDP-43 in roughly half of the neurons, suggesting that this system can also be used to study effects on untagged endogenous TDP-43 as well as TDP-43-GFP fusion protein.

Strengths:
A clearer understanding of how TDP-43 mislocalization alters cellular function, as well as pathways that mitigate clearance of TDP-43 aggregates, is critical. But modeling TDP-43 mislocalization in disease-relevant cellular systems has proven to be challenging. High levels of overexpression of TDP-43 lacking an NES can drive endogenous TDP-43 mislocalization, but such overexpression has direct and artificial consequences on certain cellular features (e.g. altered exon skipping) not seen in diseased patients. Toxic small molecules such as MG132 and arsenite can induce TDP-43 mislocalization, but co-induce myriad additional cellular dysfunctions unrelated to TDP-43 or ALS. TDP-43 binding oligonucleotides can cause cytosolic mislocalization as well. Each system has pros and cons, and additional ways to induce TDP-43 mislocalization would be useful for the field. The method described in this manuscript could provide researchers with a powerful way to study the combined biology of cytosolic TDP-43 mislocalization and nuclear TDP-43 depletion, with additional temporal control that is lacking in current method. Indeed, the authors see some evidence of differences in RNA splicing caused by pure TDP-43 depletion versus their induced mislocalization model. Finally, their method may be especially useful in determining how TDP-43 aggregates are cleared by cells, potentially revealing new biological pathways that could be therapeutically targeted.

Weaknesses:
The method and supporting data have limitations in its current form, outlined below, and in its current form the findings are rather preliminary.

• Tagging of TDP-43 with a bulky GFP tag may alter its normal physiological functions, for example phase separation properties and functions within complex ribonucleoprotein complexes. In addition, alternative isoforms of TDP-43 (e.g. "short" TDP-43, would not be GFP tagged and therefore these species would not be directly manipulatable or visualizable with the tools currently employed in the manuscript.
• The data regarding potential mislocalization of endogenous TDP-43 in the heterozygous TDP-43-GFP lines is especially intriguing and important, yet very little characterization was done. Does untagged TDP-43 co-aggregate with the tagged TDP-43? Is localization of TDP-43 immunostaining the same as the GFP signal in these cells?
• The experiments in which dox was used to induce the nanobody-NES, then dox withdrawn to study potential longer-lasting or self-perpetuating inductions of aggregation is potentially interesting. However, the nanobody was only measured at the RNA level. We know that protein half lives can be very long in neurons, and therefore residual nanobody could be present at these delayed time points. The key measurement to make would be at the protein level of the nanobody if any conclusions are be made from this experiment.
• Potential differences in splicing and microRNAs between TDP-43 knockdown and TDP-43 mislocalization are potentially interesting. However, different patterns of dysregulated RNA splicing can occur at different levels of TDP-knockdown, thus it is difficult to asses whether the changes observed in this paper are due to mislocalization per se, or rather just reflect differences in nuclear TDP-43 abundance.

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