Abstract
Background:
Inhibition of HDAC6 has been proposed as a broadly applicable therapeutic strategy for Charcot-Marie-Tooth disease (CMT). Inhibition of HDAC6 increases the acetylation of proteins important in axonal trafficking, such as alpha-tubulin and Miro, and has been shown to be efficacious in several preclinical studies using mouse models of CMT.
Aims:
Here we sought to expand on previous preclinical studies by testing the effect of genetic deletion of Hdac6 on mice carrying a humanized knockin allele of Gars1, a model of CMT type 2D.
Methods:
Gars1ΔETAQ mice were bred to an Hdac6 knockout strain, and the resulting offspring were evaluated for clinically relevant outcomes.
Results:
The genetic deletion of Hdac6 increased alpha-tubulin acetylation in the sciatic nerves of both wild type and Gars1ΔETAQ mice. However, when tested at five-weeks-of-age, the Gars1ΔETAQ mice lacking Hdac6 showed no changes in body weight, muscle atrophy, grip strength or endurance, sciatic motor nerve conduction velocity, compound muscle action potential amplitude, or peripheral nerve histopathology compared to Gars1ΔETAQ mice with intact Hdac6.
Interpretation:
Our results differ from those of two previous studies that demonstrated benefit of the HDAC6 inhibitor Tubastatin A in mouse models of CMT2D. While we cannot fully explain the different outcomes, our results offer a counterexample to the benefit of inhibiting HDAC6 in CMT2D, suggesting additional research is necessary.
Keywords: HDAC6 inhibitor, preclinical study, Charcot-Marie-Tooth disease, mouse model, tRNA synthetase
Introduction:
Charcot-Marie-Tooth disease (CMT) comprises a diverse collection of hereditary sensory and motor neuropathies (Saporta and Shy, 2013). Cumulatively, CMT affects approximately 1 in 2500 people, but mutations in at least 100 different genes in the human genome can cause CMT (Laura, et al., 2019; Skre, 1974; Timmerman, et al., 2014). Therefore, after a few common subtypes are accounted for, such as CMT1A, CMT2A, and CMT1X, individual subtypes become quite rare (DiVincenzo, et al., 2014). In addition to this genetic diversity, there are also differences in the pathophysiology and clinical diagnostic distinctions. Some forms of CMT primarily affect peripheral myelinating Schwann cells (type 1, or demyelinating CMTs), leading to reduced nerve conduction velocities, whereas other forms lead to degeneration of the peripheral sensory and motor axons themselves (type 2, or axonal CMTs), often due to specific deficits in neuronal cell biology. Nonetheless, CMT patients have shared clinical issues, including weakness, fatigue, and muscle atrophy, as well as sensory deficits, which are typically most pronounced in the distal extremities, indicating a length dependent neuropathy (Saporta and Shy, 2013).
The 100+ genes associated with CMT encode proteins involved in numerous cellular activities, such as mitochondrial function, axonal transport, endosome/lysosome trafficking, and protein synthesis (Fridman, et al., 2015). These diverse cellular functions likely indicate that there are many paths to peripheral axon dysfunction and degeneration, which should not be surprising given the extraordinary specialization of these very long cell processes. This also calls into question whether there will be a single therapy that is effective across all or many CMT subtypes. However, even if therapies are not addressing the core function of the mutated gene, there may be generally beneficial approaches that could apply to multiple forms of CMT. Such broadly beneficial approaches include the expression of neurotrophins in the periphery to maintain axon integrity (Sahenk and Ozes, 2020), the inhibition of SARM1 to prevent axon degeneration (Moss and Hoke, 2020), or the inhibition of HDAC6 to enhance alpha-tubulin acetylation and axonal transport (Rossaert and Van Den Bosch, 2020).
HDAC6 is an atypical deacetylase. The protein contains two catalytic domains and is localized primarily in the cytoplasm where it modifies proteins like alpha-tubulin and MIRO, an adapter protein for mitochondrial transport (Grozinger, et al., 1999; Hubbert, et al., 2002; Kalinski, et al., 2019; Verdel and Khochbin, 1999). The rationale for targeting HDAC6 in diseases such as CMT is that as axons become compromised, tubulin acetylation decreases. This impairs axonal transport, compromising the distal axon even more and setting up a positive feedback loop. Inhibiting HDAC6 decreases tubulin deacetylation (i.e. increases tubulin acetylation) and therefore slows this process. Improving the acetylation of MIRO could be similarly beneficial, improving mitochondrial transport in axons (Kalinski, et al., 2019). Thus, even though HDAC6 inhibitors are likely targeting a downstream consequence of diseases such as CMT, there is a basis for their mechanism of action in CMT and possibly other acquired neuropathies, such as diabetic neuropathy, for which a clinical trial has been performed (NCT03176472, clinicaltrials.gov), or even diseases such as ALS.
Inhibition of HDAC6 has been shown to be beneficial in several mouse models of CMT, including Hspb1/CMT2F, Mfn2/CMT2A, PMP22/CMT1A and Gars1/CMT2D (Benoy, et al., 2018; d’Ydewalle, et al., 2011; Ha, et al., 2020; Mo, et al., 2018; Picci, et al., 2020). In studies of Mfn2/CMT2A mice, similar results were obtained with pharmacological inhibition and genetic deletion of Hdac6 (Picci, et al., 2020). Here we focus on Gars1/CMT2D, which has been the subject of more studies than the rest. Dominant mutations in glycyl-tRNA synthetase (GARS1) cause peripheral neuropathy in both humans and mice (Antonellis, et al., 2003; Seburn, et al., 2006). Interestingly, dominant mutations in at least 5 other tRNA synthetase genes also cause forms of CMT (He, et al., 2023; Jordanova, et al., 2006; Latour, et al., 2010; Tsai, et al., 2017; Vester, et al., 2013), making the tRNA synthetases the largest gene family associated with forms of CMT and suggesting both a shared pathogenic mechanism and the possibility of shared benefit from a treatment such as HDAC6 inhibition. Two independent studies showed that treating mice with the HDAC6 inhibitor Tubastatin A mitigated the neuropathy phenotype in two mouse-specific Gars1 mutations that cause neuropathy (Benoy, et al., 2018; Mo, et al., 2018). In addition, Mo et al. showed an aberrant interaction between mutant forms of GARS1 protein and HDAC6, possibly providing a more direct molecular mechanism for this therapeutic approach (Mo, et al., 2018). Furthermore, using motor neurons derived from induced pluripotent stem cells engineered to carry a dominant GARS1 mutation, deficits in axonal transport were corrected by treatment with an HDAC6 inhibitor (Smith, et al., 2022). Taken together, these preclinical studies are encouraging regarding the potential for HDAC6 inhibitors to show benefit in CMT2D. However, here we re-examined the potential efficacy of HDAC6 inhibitors using a different strategy in which we crossed Hdac6 knockout mice to a humanized mouse model of CMT2D that carries a severe, early-onset patient allele of GARS1, the ΔETAQ allele (Morelli, et al., 2019). We find that genetic deletion of Hdac6 in the Gars1ΔETAQ background does not change the onset or severity of neuropathy, offering a counterexample to previous studies.
Materials and Methods:
Mice:
All animal experiments were conducted in compliance with the Guide on the Care and Use of Laboratory Animals and were reviewed and approved by the Institutional Animal Care and Use Committee of The Jackson Laboratory. The Hdac6 knockout strain was produced at JAX and is officially designated C57BL/6J-Hdac6em2Lutzy/J, strain #029318, MGI: 6118965. This allele deletes exons 2–4, with exon 2 containing the translation initiation codon. In internal validation studies, the mice were shown to not produce protein by western blot analysis of multiple tissues, including brain, liver, spleen, and cardiac and skeletal muscle. This result has been independently reproduced in at least three additional publications (Osseni, et al., 2020; Wang, et al., 2020a; Wang, et al., 2020b). The Gars1ΔETAQ mice have been previously described (Morelli, et al., 2019), and are officially designated Gars1em2Rwb, MGI: 6393282. Mice were maintained under standard housing conditions with food and water provided ad libitum and a 14:10 light:dark cycle.
Immunoassay:
Sciatic nerves were collected from 5-week-old mice and snap frozen. Samples were then thawed on ice, rinsed with cold PBS, and homogenized in 200 μL of lysis buffer consisting of chilled T-Per (Thermo Scientific 78510) containing Halt protease and phosphatase inhibitor cocktail (1X, Thermo Scientific 78442) and deacetylase inhibitor cocktail (1X, APExBIO K1017) in 0.5mL Dounce homogenizers. Once homogenized, lysate was transferred to an Eppendorf tube and centrifuged at max speed at 4°C for 15 minutes. Supernatant was kept and used for BCA Assay per the kit protocol (Pierce BCA Protein Assay Kit, Thermo Scientific PI23227).
Capillary immunoassays were performed using the Jess Simple Western system following the manufacturer’s protocols. Samples were prepared at 0.1 mg/ml final concentration and run on 12–230 kDa Separation Modules (Bio-Techne SM-W004). Detection was performed using the Anti-Rabbit Detection Module (Bio-Techne DM-001) and Total Protein Detection Module (Bio-Techne DM-TP01). Antibodies used were rabbit anti-alpha-tubulin (Cell Signaling Technology #2125; 1:1000), rabbit anti-acetyl-alpha-tubulin (Cell Signaling Technology #5335; 1:5000), rabbit anti-GAPDH (Cell Signaling Technology #2118; 1:1000) and the system control primary antibody (rabbit, Bio-Techne 042–196). Analysis was based on the manufacturer’s protocol, and in pilot experiments GAPDH normalization yielded similar and more consistent results than total protein, so all results presented here use GAPDH normalization. Ratios of protein concentration were then calculated using Microsoft Excel and data were plotted for visualization using GraphPad Prism.
Wire hang:
As a behavioral measure of grip strength and endurance, we utilized the inverted wire hang test (Gomez, et al., 1997; Rafael, et al., 2000; Spaulding, et al., 2016). In this assay, mice were placed on an inverted wire grid for a maximum trial time of one minute, and latency for mice to fall was measured. Mice were tested in three successive trials with a 30- to 60-second break in between trials and the average time-to-fall was reported. Mice were not trained prior to the initial test session.
Neurophysiology:
Nerve conduction velocity of motor axons of the sciatic nerve was measured (Burgess, et al., 2010; Morelli, et al., 2017). Mice were anaesthetized with 2% isoflurane and placed on a thermostatically regulated heating pad to maintain body temperature. Action potentials were produced by placing stimuli proximally at the sciatic notch or distally at the ankle. The latencies to elicit a compound muscle action potential (CMAP) recorded in the thenar muscles of the hind paw following proximal and distal stimuli were determined. NCV was calculated as [conduction distance/(proximal latency-distal latency)]. Amplitude of the CMAP was also measured.
Histopathology:
Nerve histology was performed as previously described (Burgess, et al., 2010). In brief, the femoral motor and sensory nerves were fixed in place using a drop of 2% paraformaldehyde, 2% glutaraldehyde in a 0.1M cacodylate buffer. The nerves were then dissected free, post-fixed overnight in the same solution and rinsed into PBS. Samples were dehydrated and plastic embedded for sectioning. Thick sections (0.5 μm) were placed on slides and stained with toluidine blue for light microscopy. Images were collected on a Nikon Eclipse 600 microscope with a 40X objective. Axon number and size were quantified using ImageJ.
Statistics:
Statistical tests were performed using GraphPad Prism software. Comparisons across four genotypes were tested using an ordinary one-way ANOVA with multiple comparisons and lack of significance was confirmed in pairwise comparisons using a Student’s t test. Differences in the cumulative frequency distribution of axon areas were tested using nested one-way ANOVA with multiple comparisons and lack of significance was confirmed in pairwise comparisons using a nested t test. Differences were considered significant with a p value < 0.05.
Results:
The Hdac6 gene is on the X chromosome in mice. Therefore, we bred a Gars1ΔETAQ male to a heterozygous Hdac6 knockout (Hdac6KO/+) female. The Hdac6ko mice carry a deletion of exons 2–4 of the Hdac6 gene and do not produce detectable protein, as shown previously (Osseni, et al., 2020; Wang, et al., 2020a; Wang, et al., 2020b). We analyzed male offspring of this cross, half of which carried the dominant Gars1 mutation and half of which carried the Hdac6 knockout allele. The Gars1ΔETAQ mice have an early onset peripheral neuropathy, with changes evident by a few weeks of age (Morelli, et al., 2019). We therefore performed all analyses at 5 weeks of age. To evaluate the effect of the Hdac6 knockout on alpha-tubulin acetylation, we used a digital capillary western blot to compare total alpha-tubulin and acetylated alpha-tubulin in the sciatic nerves of mice of all four genotypes [1) WT;WT, 2) WT;Hdac6KO, 3) Gars1ΔETAQ;WT, and 4) Gars1ΔETAQ;Hdac6KO] using GAPDH as an internal standard (Figure 1). In both wild type and Gars1ΔETAQ backgrounds, the loss of Hdac6 led to a significant (~2 fold) increase in acetylated alpha-tubulin. This was anticipated and indicates that our genetic approach does indeed increase the acetylation of HDAC6 targets in peripheral nerve, although we do not understand the increased variability in tubulin acetylation levels in the Hdac6KO background compared to the tightly consistent levels seen in wild-type and Gars1ΔETAQ mice. The Gars1ΔETAQ samples did not show a significant decrease in acetylated alpha-tubulin compared to wild-type when tested by ANOVA including all four genotypes, despite this result being shown previously (Benoy, et al., 2018; Mo, et al., 2018; Smith, et al., 2022). However, using a t test in a pairwise comparison of Gars1ΔETAQ;WT vs WT;WT yielded a statistically significant result (16.5 ± 4.3% decrease in Gars1ΔETAQ, p=0.003). A stronger decrease in alpha-tubulin acetylation the Gars1ΔETAQ mice may have been found if we had examined older mice or more distal nerves than the sciatic. Previous studies suggest that GARS1 protein levels are unchanged or perhaps slightly elevated in the dominant mutant alleles (see for example (Motley, et al., 2011)); however, here we did not confirm whether GARS1 levels change in the absence of Hdac6.
Figure 1:

Digital western blotting of alpha tubulin and acetylated alpha-tubulin. Sciatic nerve samples from mice of the indicated genotypes (n=6 per group) were analyzed by digital western for the ratio of acetylated to total tubulin, normalized to GAPDH. Data were analyzed by ordinary one-way ANOVA with Tukey’s multiple comparisons test. The more sensitive t-test was also applied to the pairwise comparison Gars1ΔETAQ;WT vs WT;WT, yielding a statistically significant result (p=0.003).
Given that our analyses include four genotypes, we used one-way ANOVA to test for significance, as in the results presented in figure 1. As described above, however, the more sensitive t-test was needed to detect a significant decrease in tubulin acetylation in Gars1ΔETAQ sciatic nerve. In all results presented in figures 2–4, the Gars1+/+ and Gars1ΔETAQ mice differed significantly when analyzed by ANOVA, but the Hdac6 genotype had no effect in either the Gars1+/+ or Gars1ΔETAQ mice. To perform a more targeted pairwise comparison, we also used a t test to ask whether Gars1ΔETAQ mice differed with and without the Hdac6KO allele. Nested ANOVA and t test were used for the axon area distributions in figure 4C. We did not find differences between Gars1ΔETAQ and Gars1ΔETAQ without Hdac6, even using the pairwise t tests.
Figure 2:

Neuromuscular phenotype of Gars1;Hdac6 mice. A) Body weight, B) muscle weight to body weight ratio (a measure of muscle atrophy), and C) motor performance as assayed by the average latency to fall from an inverted wire grid. For all measures, Gars1ΔETAQ mice had values that were significantly less than those of Gars1WT mice, regardless of Hdac6 genotype. In contrast, genetic ablation of Hdac6 had no effect in Gars1ΔETAQ on any measure, even when the more sensitive t test was used to compare Gars1ΔETAQ mice with and without Hdac6. Data are shown as mean ± S.D. with points representing individual mice with the following numbers per group: WT=10, Hdac6KO=11, Gars1ΔETAQ=15 and Gars1ΔETAQ;Hdac6KO=17.
Figure 4:

Femoral motor nerve histopathology of Gars1;Hdac6 mice. A) Images of cross-sections of femoral motor nerve, showing the smaller overall nerve area of Gars1ΔETAQ mice, regardless of Hdac6 genotype. B) Number of myelinated axons in the motor branch of the femoral nerve. The Gars1ΔETAQ mutation causes a decrease in the number of axons, and Hdac6 ablation has no effect. C) Area of myelinated axons in the motor branch of the femoral nerve, shown by the cumulative frequency distribution. The Gars1ΔETAQ mutation causes a decrease in axon area, and Hdac6 ablation has no effect. Data in (B) are shown as mean ± S.D. with points representing individual mice with the following numbers per group: WT=10, Hdac6KO=11, Gars1ΔETAQ=15 and Gars1ΔETAQ;Hdac6KO=17. Scale bar in (A) = 50μm for all images.
The Gars1ΔETAQ mice have a reduced body weight by 5 weeks-of-age, and this was not affected by the loss of Hdac6 (Figure 2A). The reduced body weight is, at least in part, attributed to muscle atrophy. This is reflected in a reduction in the ratio of the weight of the triceps surae muscles to total body weight (MW:BW ratio). This measure of muscle atrophy also was not altered by the loss of Hdac6 (Figure 2B). We also used the wire hang test to assess grip strength and endurance. The Gars1ΔETAQ perform very poorly in this test and hang on less than 20 seconds before falling, whereas wild type mice typically hang on for the full one-minute duration of the test. The deletion of Hdac6 did not improve the performance of the Gars1ΔETAQ mice in this task (Figure 2C).
We also assessed the effects of deleting Hdac6 using neurophysiology. The sciatic motor nerve conduction velocity is markedly reduced in the Gars1ΔETAQ mice, as we have seen in other Gars1/CMT2D mouse models (Achilli, et al., 2009; Seburn, et al., 2006). The genetic deletion of Hdac6 did not improve nerve conduction velocity in the Gars1ΔETAQ mice (Figure 3A). In contrast, other treatments such as inhibiting GCN2 and the integrated stress response or allele-specific knockdown of mutant Gars1 mRNA using gene therapy approaches were able to significantly improve this outcome (Morelli, et al., 2019; Spaulding, et al., 2021). The amplitude of the compound muscle action potential in the thenar muscles of the foot following stimulation of the nerve at the ankle is also reduced in the Gars1ΔETAQ mice, consistent with denervation and/or failures in synaptic transmission at the neuromuscular junction. Deletion of Hdac6 had no effect on CMAP amplitude (Figure 3B).
Figure 3:

Neurophysiology of Gars1;Hdac6 mice. A) Sciatic motor nerve conduction velocity and B) compound muscle action potential amplitude were both decreased in Gars1ΔETAQ mice compared to Gars1WT, regardless of Hdac6 genotype. As in Figure 2, genetic ablation of Hdac6 had no effect in Gars1ΔETAQ on either measure, even when the more sensitive t test was used. Data are shown as mean ± S.D. with points representing individual mice with the following numbers per group : WT=10, Hdac6KO=11, Gars1ΔETAQ=15 and Gars1ΔETAQ;Hdac6KO=17.
Finally, we examined peripheral nerves by histopathology. The motor branch of the femoral nerve, which innervates the quadriceps muscles of the thigh, shows loss of myelinated axons in the Gars1ΔETAQ mice, and the remaining axons are smaller. Neither axon number nor axon size was improved with the deletion of Hdac6 (Figure 4A–C).
Discussion:
In this study, we revisited the potential benefit of inhibiting HDAC6 as a treatment for CMT2D, caused by dominant GARS1 mutations. We used an approach that differs from previous studies in two major ways. First, we used genetic deletion of Hdac6 on the X chromosome of mice instead of using a pharmacological inhibitor such as Tubastatin A. Second, we used a mouse model of CMT2D carrying a small internal deletion in exon 8 of GARS1 that was identified in a patient with severe early onset motor neuropathy (Morelli, et al., 2019). Previous studies used alleles of mouse Gars1 that cause peripheral neuropathy, but the amino acids changed (P278KY and C201R) are different than known human disease variants (Benoy, et al., 2018; Mo, et al., 2018). However, in other studies, the Gars1ΔETAQ mice have behaved very similarly to other alleles (Morelli, et al., 2019; Spaulding, et al., 2021). Unlike previous studies (Benoy, et al., 2018; Mo, et al., 2018), we found no benefit from eliminating Hdac6 in the Gars1ΔETAQ background by any of our outcome measures (body weight, muscle atrophy, motor performance, sciatic motor nerve conduction velocity, compound muscle action potential amplitude, and peripheral nerve histopathology, figures 2–4) despite robust elevation of acetylation of alpha-tubulin in peripheral nerves following Hdac6 deletion (figure 1). Below, we will discuss differences in these experiments and possible interpretations.
In the study by Mo et al., Gars1P278KY (also called P234KY due to differences caused by the inclusion/exclusion of a mitochondrial targeting sequence in the amino acid count) were used (Mo, et al., 2018). These mice are similar to, but slightly more severe than, the Gars1ΔETAQ mice. Decreases in alpha-tubulin acetylation (~3-fold) were seen as early as one week postnatally in peripheral nerves of Gars1P278KY mice, while we saw smaller decreases in alpha-tubulin acetylation in the Gars1ΔETAQ mice compared to wild-type controls at 5 weeks of age. This may reflect a more pronounced change in tubulin acetylation in the P278KY allele, and we may have found more pronounced changes in acetylation if we have aged the ΔETAQ allele longer, or had looked in more distal nerves, as seen in Mfn2 mice modeling CMT2A, for example (Picci, et al., 2020). In Mo et al., axonal transport of quantum-dot labeled Nerve Growth Factor (NGF) was decreased in cultured DRG neurons from the Gars1P278KY mice, and this was corrected by treatment with the HDAC6 inhibitor Tubastatin A (Mo, et al., 2018). Similar results for mitochondrial transport have been reported in human iPSC-derived neurons engineered to carry the same mutation (Smith, et al., 2022). Additionally, retrograde axonal transport of signaling endosomes labeled with a fluorescent tetanus toxin heavy chain is restored in vivo in Gars1/CMT2D mice by other methods such as treatment with the neurotrophin BDNF (Sleigh, et al., 2023). We did not evaluate axonal transport in this study, and while the improvements in axonal transport observed in multiple previous studies are encouraging (Mo, et al., 2018; Sleigh, et al., 2023; Smith, et al., 2022), improved axonal transport has yet to be causally tied to improved clinically relevant outcomes. In in vivo studies, treatment of the Gars1P278KY mice with Tubastatin A starting at 35 days of age and lasting 2 weeks led to a modest improvement in hind limb clasping, rotarod performance, and gait parameters, but the mice still performed well below wild type littermates (Mo, et al., 2018). When the same Gars1P278KY mice are bred to a Gcn2 knockout strain, the double mutants perform comparably to littermate controls (Spaulding, et al., 2021), so the effects of Tubastatin A are comparatively modest, but Tubastatin A treatment was started after the onset of a phenotype and only lasted two weeks, so the comparison is complicated.
The study by Benoy et al. reached similar conclusions to Mo et al. The Benoy et al. study used Gars1C201R mice, which have a milder phenotype than either ΔETAQ or P278KY (Benoy, et al., 2018). These mice also showed decreased alpha-tubulin acetylation selectively in sciatic nerve, but this was assayed in one year old animals, consistent with the idea that here we may have seen greater changes in tubulin acetylation in the ΔETAQ mice in older animals. Decreased axonal transport of mitochondria in cultured DRG neurons was again observed, and was corrected by Tubastatin A. In in vivo studies, the Gars1C201R mice were treated with Tubastatin A at 4 months of age for 40 days. This led to an improvement in grip strength, performance in a turning grid test, CMAP amplitude, sensory nerve action potential (SNAP) amplitude and innervation at the neuromuscular junction. Rotarod performance and performance in the wire hang test were not significantly improved. The values for Gars1C201R mice after Tubastatin A treatment are still well below those of control mice reported previously in the same study, so like Mo et al., the effect size was modest.
In our study, we saw no improvement with the genetic deletion of Hdac6. Our studies were well-powered, with 15 Gars1ΔETAQ mice compared to 17 double Gars1ΔETAQ:Hdac6KO mutants, for example. Our outcome measures were largely quantitative values that did not depend on subjective judgement from the tester and these outcomes have been responsive to treatment in other preclinical studies (Morelli, et al., 2019; Spaulding, et al., 2021). The tester was not technically blinded to genotype during data collection or analysis, but this lack of effect was also not the anticipated result when these studies were initiated, given the previous results with Tubastatin A in other mouse models of CMT2D. Some of our tests were the same as those used by Benoy et al., including wire hang (no improvement in either study) and CMAP amplitude (improved in Benoy et al., but not here). Similarly, Benoy et al. did not see an improvement in rotarod performance, whereas Mo et al. reported an improvement. Therefore, previous findings were also not completely consistent.
A criticism of genetic approaches such as the deletion of Hdac6 used here instead of pharmacological approaches is that the intervention is present from conception and thus is effectively started before disease onset, which is less clinically relevant. This is true, but should have worked in our favor, and instead, we saw no improvement in the Gars1ΔETAQ mice with the deletion of Hdac6. An additional criticism of genetic approaches is the possibility of compensation by related genes. As HDAC6 is an atypical deacetylase, this is less likely, and we found elevated levels of tubulin acetylation in the sciatic nerves of Hdac6KO mice (figure 1), indicating that any compensation by other deacetylases is incomplete. The only other type IIb HDAC is HDAC10, making it the most likely to compensate, and it is indeed expressed in alpha motor neurons in the mouse spinal cord, albeit at an mRNA level roughly half that of Hdca6 (https://seqseek.ninds.nih.gov/spinalcordinjury) (Alkaslasi, et al., 2021). In contrast, pharmacological inhibitors carry the possible caveat of off-target effects. Whether the benefits of Tubastatin A in CMT2D mice are due to lack of specificity for HDAC6 is unknown, although Tubastatin A has been shown to also significantly inhibit HDAC10 as well as sirtuin deacetylases, any of which may be partially redundant with and compensatory for HDAC6 (Choi, et al., 2019; Ptacek, et al., 2023). Additional tests with other HDAC6 inhibitors may clarify this point. Alternatively, inhibitors could be tested in an Hdac6KO background, where they would be anticipated to have no additional effect, to establish their specificity.
Thus, it is unclear why our genetic deletion model did not reproduce the effects reported in previous studies of Gars1/CMT2D mice that used HDAC6 inhibitors. Perhaps the effects of Hdac6 deletion would have been more pronounced on disease progression and we would have observed differences if we had aged the animals longer, but Mo et al. reported effects after just two weeks of treatment and Benoy et al. after 40 days. Our results do clearly indicate that the reported interaction between mutant forms of GARS1 protein and HDAC6 is not a causal part of the disease mechanism, as the complete removal of Hdac6 did not alter the disease course at our five-week time point. Our results offer a counterexample to previous studies on the efficacy of inhibiting HDAC6 as a treatment for CMT2D and suggest that additional research is necessary to validate HDAC6 as a therapeutic target for this disease and to clarify the magnitude of the anticipated benefit.
Acknowledgements:
The authors would like to thank the Scientific Services at The Jackson Laboratory for their support of this project, particularly Mr. Pete Finger for assistance with nerve histopathology. The Scientific Services are supported by NIH grant CA34196. This work was funded by NIH (R37NS054154 to RWB) and the Charcot-Marie-Tooth Association.
Footnotes
Conflict of Interest: The authors declare no conflicts of interest. Dr. Burgess has two pending patents, 17/271,377 2021 and 17/288,178 2021, and has been a paid consultant for Roche. These interests do not influence the work presented here.
Data availability:
All data relevant to this study are presented in the paper. Raw data are available upon reasonable request by contacting the authors.
References:
- Achilli F, Bros-Facer V, Williams HP, Banks GT, AlQatari M, Chia R, Tucci V, Groves M, Nickols CD, Seburn KL, Kendall R, Cader MZ, Talbot K, van Minnen J, Burgess RW, Brandner S, Martin JE, Koltzenburg M, Greensmith L, Nolan PM, Fisher EM (2009). An ENU-induced mutation in mouse glycyl-tRNA synthetase (GARS) causes peripheral sensory and motor phenotypes creating a model of Charcot-Marie-Tooth type 2D peripheral neuropathy. Disease models & mechanisms 2:359–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alkaslasi MR, Piccus ZE, Hareendran S, Silberberg H, Chen L, Zhang Y, Petros TJ, Le Pichon CE (2021). Single nucleus RNA-sequencing defines unexpected diversity of cholinergic neuron types in the adult mouse spinal cord. Nature communications 12:2471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Antonellis A, Ellsworth RE, Sambuughin N, Puls I, Abel A, Lee-Lin SQ, Jordanova A, Kremensky I, Christodoulou K, Middleton LT, Sivakumar K, Ionasescu V, Funalot B, Vance JM, Goldfarb LG, Fischbeck KH, Green ED (2003). Glycyl tRNA synthetase mutations in Charcot-Marie-Tooth disease type 2D and distal spinal muscular atrophy type V. American journal of human genetics 72:1293–1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benoy V, Van Helleputte L, Prior R, d’Ydewalle C, Haeck W, Geens N, Scheveneels W, Schevenels B, Cader MZ, Talbot K, Kozikowski AP, Vanden Berghe P, Van Damme P, Robberecht W, Van Den Bosch L (2018). HDAC6 is a therapeutic target in mutant GARS-induced Charcot-Marie-Tooth disease. Brain : a journal of neurology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burgess RW, Cox GA, Seburn KL (2010). Neuromuscular disease models and analysis. Methods in molecular biology 602:347–393. [DOI] [PubMed] [Google Scholar]
- Choi YJ, Kang MH, Hong K, Kim JH (2019). Tubastatin A inhibits HDAC and Sirtuin activity rather than being a HDAC6-specific inhibitor in mouse oocytes. Aging (Albany NY) 11:1759–1777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- d’Ydewalle C, Krishnan J, Chiheb DM, Van Damme P, Irobi J, Kozikowski AP, Vanden Berghe P, Timmerman V, Robberecht W, Van Den Bosch L (2011). HDAC6 inhibitors reverse axonal loss in a mouse model of mutant HSPB1-induced Charcot-Marie-Tooth disease. Nat Med 17:968–974. [DOI] [PubMed] [Google Scholar]
- DiVincenzo C, Elzinga CD, Medeiros AC, Karbassi I, Jones JR, Evans MC, Braastad CD, Bishop CM, Jaremko M, Wang Z, Liaquat K, Hoffman CA, York MD, Batish SD, Lupski JR, Higgins JJ (2014). The allelic spectrum of Charcot-Marie-Tooth disease in over 17,000 individuals with neuropathy. Molecular genetics & genomic medicine 2:522–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fridman V, Bundy B, Reilly MM, Pareyson D, Bacon C, Burns J, Day J, Feely S, Finkel RS, Grider T, Kirk CA, Herrmann DN, Laura M, Li J, Lloyd T, Sumner CJ, Muntoni F, Piscosquito G, Ramchandren S, Shy R, Siskind CE, Yum SW, Moroni I, Pagliano E, Zuchner S, Scherer SS, Shy ME, Inherited Neuropathies C (2015). CMT subtypes and disease burden in patients enrolled in the Inherited Neuropathies Consortium natural history study: a cross-sectional analysis. Journal of neurology, neurosurgery, and psychiatry 86:873–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gomez CM, Maselli R, Gundeck JE, Chao M, Day JW, Tamamizu S, Lasalde JA, McNamee M, Wollmann RL (1997). Slow-channel transgenic mice: a model of postsynaptic organellar degeneration at the neuromuscular junction. J Neurosci 17:4170–4179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grozinger CM, Hassig CA, Schreiber SL (1999). Three proteins define a class of human histone deacetylases related to yeast Hda1p. Proceedings of the National Academy of Sciences of the United States of America 96:4868–4873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ha N, Choi YI, Jung N, Song JY, Bae DK, Kim MC, Lee YJ, Song H, Kwak G, Jeong S, Park S, Nam SH, Jung SC, Choi BO (2020). A novel histone deacetylase 6 inhibitor improves myelination of Schwann cells in a model of Charcot-Marie-Tooth disease type 1A. British journal of pharmacology 177:5096–5113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He J, Liu XX, Ma MM, Lin JJ, Fu J, Chen YK, Xu GR, Xu LQ, Fu ZF, Xu D, Chen WF, Cao CY, Shi Y, Zeng YH, Zhang J, Chen XC, Zhang RX, Wang N, Kennerson M, Fan DS, Chen WJ (2023). Heterozygous Seryl-tRNA Synthetase 1 Variants Cause Charcot-Marie-Tooth Disease. Annals of neurology 93:244–256. [DOI] [PubMed] [Google Scholar]
- Hubbert C, Guardiola A, Shao R, Kawaguchi Y, Ito A, Nixon A, Yoshida M, Wang XF, Yao TP (2002). HDAC6 is a microtubule-associated deacetylase. Nature 417:455–458. [DOI] [PubMed] [Google Scholar]
- Jordanova A, Irobi J, Thomas FP, Van Dijck P, Meerschaert K, Dewil M, Dierick I, Jacobs A, De Vriendt E, Guergueltcheva V, Rao CV, Tournev I, Gondim FA, D’Hooghe M, Van Gerwen V, Callaerts P, Van Den Bosch L, Timmermans JP, Robberecht W, Gettemans J, Thevelein JM, De Jonghe P, Kremensky I, Timmerman V (2006). Disrupted function and axonal distribution of mutant tyrosyl-tRNA synthetase in dominant intermediate Charcot-Marie-Tooth neuropathy. Nature genetics 38:197–202. [DOI] [PubMed] [Google Scholar]
- Kalinski AL, Kar AN, Craver J, Tosolini AP, Sleigh JN, Lee SJ, Hawthorne A, Brito-Vargas P, Miller-Randolph S, Passino R, Shi L, Wong VSC, Picci C, Smith DS, Willis DE, Havton LA, Schiavo G, Giger RJ, Langley B, Twiss JL (2019). Deacetylation of Miro1 by HDAC6 blocks mitochondrial transport and mediates axon growth inhibition. The Journal of cell biology 218:1871–1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Latour P, Thauvin-Robinet C, Baudelet-Mery C, Soichot P, Cusin V, Faivre L, Locatelli MC, Mayencon M, Sarcey A, Broussolle E, Camu W, David A, Rousson R (2010). A Major Determinant for Binding and Aminoacylation of tRNA(Ala) in Cytoplasmic Alanyl-tRNA Synthetase Is Mutated in Dominant Axonal Charcot-Marie-Tooth Disease. American journal of human genetics 86:77–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laura M, Pipis M, Rossor AM, Reilly MM (2019). Charcot-Marie-Tooth disease and related disorders: an evolving landscape. Current opinion in neurology 32:641–650. [DOI] [PubMed] [Google Scholar]
- Mo Z, Zhao X, Liu H, Hu Q, Chen XQ, Pham J, Wei N, Liu Z, Zhou J, Burgess RW, Pfaff SL, Caskey CT, Wu C, Bai G, Yang XL (2018). Aberrant GlyRS-HDAC6 interaction linked to axonal transport deficits in Charcot-Marie-Tooth neuropathy. Nature communications 9:1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morelli KH, Griffin LB, Pyne NK, Wallace LM, Fowler AM, Oprescu SN, Takase R, Wei N, Meyer-Schuman R, Mellacheruvu D, Kitzman JO, Kocen SG, Hines TJ, Spaulding EL, Lupski JR, Nesvizhskii A, Mancias P, Butler IJ, Yang XL, Hou YM, Antonellis A, Harper SQ, Burgess RW (2019). Allele-specific RNA interference prevents neuropathy in Charcot-Marie-Tooth disease type 2D mouse models. J Clin Invest 129:5568–5583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morelli KH, Seburn KL, Schroeder DG, Spaulding EL, Dionne LA, Cox GA, Burgess RW (2017). Severity of Demyelinating and Axonal Neuropathy Mouse Models Is Modified by Genes Affecting Structure and Function of Peripheral Nodes. Cell reports 18:3178–3191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moss KR, Hoke A (2020). Targeting the programmed axon degeneration pathway as a potential therapeutic for Charcot-Marie-Tooth disease. Brain research 1727:146539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Motley WW, Seburn KL, Nawaz MH, Miers KE, Cheng J, Antonellis A, Green ED, Talbot K, Yang XL, Fischbeck KH, Burgess RW (2011). Charcot-Marie-Tooth-Linked Mutant GARS Is Toxic to Peripheral Neurons Independent of Wild-Type GARS Levels. PLoS Genet 7:e1002399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Osseni A, Ravel-Chapuis A, Thomas JL, Gache V, Schaeffer L, Jasmin BJ (2020). HDAC6 regulates microtubule stability and clustering of AChRs at neuromuscular junctions. The Journal of cell biology 219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Picci C, Wong VSC, Costa CJ, McKinnon MC, Goldberg DC, Swift M, Alam NM, Prusky GT, Shen S, Kozikowski AP, Willis DE, Langley B (2020). HDAC6 inhibition promotes alpha-tubulin acetylation and ameliorates CMT2A peripheral neuropathy in mice. Experimental neurology 328:113281. [DOI] [PubMed] [Google Scholar]
- Ptacek J, Snajdr I, Schimer J, Kutil Z, Mikesova J, Baranova P, Havlinova B, Tueckmantel W, Majer P, Kozikowski A, Barinka C (2023). Selectivity of Hydroxamate- and Difluoromethyloxadiazole-Based Inhibitors of Histone Deacetylase 6 In Vitro and in Cells. Int J Mol Sci 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rafael JA, Nitta Y, Peters J, Davies KE (2000). Testing of SHIRPA, a mouse phenotypic assessment protocol, on Dmd(mdx) and Dmd(mdx3cv) dystrophin-deficient mice. Mamm Genome 11:725–728. [DOI] [PubMed] [Google Scholar]
- Rossaert E, Van Den Bosch L (2020). HDAC6 inhibitors: Translating genetic and molecular insights into a therapy for axonal CMT. Brain research 1733:146692. [DOI] [PubMed] [Google Scholar]
- Sahenk Z, Ozes B (2020). Gene therapy to promote regeneration in Charcot-Marie-Tooth disease. Brain research 1727:146533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saporta MA, Shy ME (2013). Inherited peripheral neuropathies. Neurologic clinics 31:597–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seburn KL, Nangle LA, Cox GA, Schimmel P, Burgess RW (2006). An active dominant mutation of glycyl-tRNA synthetase causes neuropathy in a Charcot-Marie-Tooth 2D mouse model. Neuron 51:715–726. [DOI] [PubMed] [Google Scholar]
- Skre H (1974). Genetic and clinical aspects of Charcot-Marie-Tooth’s disease. Clin Genet 6:98–118. [DOI] [PubMed] [Google Scholar]
- Sleigh JN, Villarroel-Campos D, Surana S, Wickenden T, Tong Y, Simkin RL, Vargas JNS, Rhymes ER, Tosolini AP, West SJ, Zhang Q, Yang XL, Schiavo G (2023). Boosting peripheral BDNF rescues impaired in vivo axonal transport in CMT2D mice. JCI Insight 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith AST, Kim JH, Chun C, Gharai A, Moon HW, Kim EY, Nam SH, Ha N, Song JY, Chung KW, Doo HM, Hesson J, Mathieu J, Bothwell M, Choi BO, Kim DH (2022). HDAC6 Inhibition Corrects Electrophysiological and Axonal Transport Deficits in a Human Stem Cell-Based Model of Charcot-Marie-Tooth Disease (Type 2D). Adv Biol (Weinh) 6:e2101308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spaulding EL, Hines TJ, Bais P, Tadenev ALD, Schneider R, Jewett D, Pattavina B, Pratt SL, Morelli KH, Stum MG, Hill DP, Gobet C, Pipis M, Reilly MM, Jennings MJ, Horvath R, Bai Y, Shy ME, Alvarez-Castelao B, Schuman EM, Bogdanik LP, Storkebaum E, Burgess RW (2021). The integrated stress response contributes to tRNA synthetase-associated peripheral neuropathy. Science 373:1156–1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spaulding EL, Sleigh JN, Morelli KH, Pinter MJ, Burgess RW, Seburn KL (2016). Synaptic Deficits at Neuromuscular Junctions in Two Mouse Models of Charcot-Marie-Tooth Type 2d. The Journal of neuroscience : the official journal of the Society for Neuroscience 36:3254–3267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Timmerman V, Strickland AV, Zuchner S (2014). Genetics of Charcot-Marie-Tooth (CMT) Disease within the Frame of the Human Genome Project Success. Genes 5:13–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai PC, Soong BW, Mademan I, Huang YH, Liu CR, Hsiao CT, Wu HT, Liu TT, Liu YT, Tseng YT, Lin KP, Yang UC, Chung KW, Choi BO, Nicholson GA, Kennerson ML, Chan CC, De Jonghe P, Cheng TH, Liao YC, Zuchner S, Baets J, Lee YC (2017). A recurrent WARS mutation is a novel cause of autosomal dominant distal hereditary motor neuropathy. Brain : a journal of neurology 140:1252–1266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verdel A, Khochbin S (1999). Identification of a new family of higher eukaryotic histone deacetylases. Coordinate expression of differentiation-dependent chromatin modifiers. The Journal of biological chemistry 274:2440–2445. [DOI] [PubMed] [Google Scholar]
- Vester A, Velez-Ruiz G, McLaughlin HM, Lupski JR, Talbot K, Vance JM, Zuchner S, Roda RH, Fischbeck KH, Biesecker LG, Nicholson G, Beg AA, Antonellis A (2013). A loss-of-function variant in the human histidyl-tRNA synthetase (HARS) gene is neurotoxic in vivo. Human mutation 34:191–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y, Wang K, Fu J (2020a). HDAC6 Mediates Macrophage iNOS Expression and Excessive Nitric Oxide Production in the Blood During Endotoxemia. Front Immunol 11:1893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y, Wang K, Fu J (2020b). HDAC6 Mediates Poly (I:C)-Induced TBK1 and Akt Phosphorylation in Macrophages. Front Immunol 11:1776. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
All data relevant to this study are presented in the paper. Raw data are available upon reasonable request by contacting the authors.
