Abstract Amyotrophic lateral sclerosis (ALS) involves motor neuron death due to mislocalized TDP-43. Pathologic TDP-43 associates with stress granules (SGs), and lowering the SG-associated protein ataxin-2 (ATXN2) using Atxn2-targeting antisense oligonucleotides prolongs survival in TAR4/4 sporadic ALS mice but failed in clinical trials likely due to poor target engagement. Here we show that an AAV with potent motor neuron transduction delivering Atxn2-targeting miRNAs reduces Atxn2 throughout the central nervous system at doses 40x lower than published work. In TAR4/4 mice, miAtxn2 increased survival (50%) and strength, and reduced motor neuron death, inflammation, and phosphorylated TDP-43. TAR4/4 transcriptomic dysregulation recapitulated ALS gene signatures that were rescued by miAtxn2, identifying potential therapeutic mechanisms and biomarkers. In slow progressing hemizygous mice, miAtxn2 slowed disease progression, and in ALS patient-derived lower motor neurons, our AAV vector transduced >95% of cells and potently reduced ATXN2 at MOI 4 logs lower than previously reported. These data support AAV-RNAi targeting ATXN2 as a translatable therapy for sporadic ALS. Subject terms: Amyotrophic lateral sclerosis, Amyotrophic lateral sclerosis, RNAi __________________________________________________________________ Amado et al. develop a gene therapy for sporadic ALS using motor neuron-targeting AAVs to deliver RNAi targeting ataxin-2. In a mouse model, survival, strength, and disease-related pathology are improved; and human motor neurons are strongly transduced. Introduction Amyotrophic lateral sclerosis (ALS) is a uniformly fatal disease characterized by progressive degeneration of upper and lower motor neurons in the cerebral motor cortex and anterior horn cells of the spinal cord, respectively. Although 10% of cases are familial and an additional 10–15% of patients have an identifiable causal mutation^[53]1,[54]2, 97% of all ALS cases have a shared downstream pathology of cytoplasmic mislocalization and aggregation of a key nuclear protein, Tar DNA binding protein of 43 kD (TDP-43). A contributory event to TDP-43 mislocalization is its aggregation in cytoplasmic stress granules, resulting in neuronal death through cytoplasmic gain-of-function^[55]3,[56]4 and nuclear loss-of-function^[57]5–[58]7. Inhibiting stress granule formation is therefore a promising therapeutic strategy. One approach to achieve this is downregulating the stress granule-associated protein ataxin-2 (ATXN2). Indeed, recent work showed that antisense oligonucleotides (ASOs) could prolong survival by 35% in a mouse model of sporadic ALS^[59]8. Disappointingly, this strategy was not successful in a recent clinical trial ([60]NCT04494256), in which no clinical improvement was seen, and neurofilament light chain (NfL), a biomarker of neurodegeneration, was not reduced in the cerebrospinal fluid (CSF). However, only 21% reduction of CSF ATXN2 was achieved, and only 12% more reduction than was seen in placebo^[61]9, an amount far lower than the anticipated reductions predicted from preclinical studies. As the maximal safe dose had been administered, further development of this strategy was halted. Therefore, the effects of ATXN2 knockdown in ALS patients remain unknown. An alternative approach is to achieve strong and lasting knockdown throughout motor neurons of the brain and spinal cord using AAV-mediated RNAi delivery. If effective, this could be an attractive alternative to ASOs, reducing the morbidity and logistical challenges associated with repeated intrathecal injection. Here, we designed and packaged microRNAs (miRNA) targeting Atxn2 into a peptide-modified AAV9 variant, PM-AAV9, engineered for superior CNS targeting, and showed that a single intracerebroventricular (ICV) injection early in life provides sustained knockdown throughout the mouse central nervous system. In the same ALS mouse model used in ASO studies^[62]8,[63]10, we demonstrate that miAtxn2 treatment markedly improves survival, strength, and motor coordination. miAtxn2 treatment also decreases TDP-43 histopathology, reduces inflammation, and improves motor neuron phenotypes. We further demonstrate correction of key ALS-associated transcripts dysregulated in this model and demonstrate the ability of our vectors to both transduce and reduce human ATXN2 levels in human lower motor neurons, supporting the potential of AAV-mediated RNAi against ATXN2 as a promising treatment strategy for the 97% of ALS cases characterized by TDP-43 pathology. Results Generation of miRNA, capsid selection, and ATXN2 knockdown efficacy MiRNAs targeting mouse Atxn2 were designed and screened for knockdown efficacy in murine N2A cells (Fig. [64]S1A) as well as optimized for strand loading bias. From this screen, miRNA V1, which targets both mouse and human transcripts, was used for further studies, hereby referred to as miAtxn2 (Fig. [65]S1B). In parallel, three different AAV9 capsid variants were assessed for transduction efficiency of upper motor neurons in the cerebral cortex and lower motor neurons in the anterior horn of the mouse spinal cord to determine the optimal serotype for miAtxn2 delivery. Pups were generated by crossing wildtype C57Bl/6 mice with wildtype SJL mice to match the mixed background of the ALS mouse model to ensure strain-relevance of the transduction results. Pups were injected at postnatal day 1 (P1) in the lateral ventricles with identical doses of either wildtype AAV9, AAV-PHP.eB, or a peptide-modified AAV9 that we developed in earlier work^[66]11, AAV9.KGG, which we here refer to as PM-AAV9. Each capsid was used to deliver enhanced GFP (eGFP) driven by a universal promoter (Fig. [67]1A). Native fluorescence visualization of brains (Fig. [68]1B, C) and lumbar spinal cord sections (Fig. [69]1D) showed that PM-AAV9 provided superior transduction, with strong expression in Layer V of the motor cortex (Fig. [70]1C) and anterior horn cells of the spinal cord (Fig. [71]1D). Fig. 1. Capsid selection and AAV-miAtxn2-mediated reduction of ataxin-2 in target cells. Fig. 1 [72]Open in a new tab A Schematic of vector and delivery route used in capsid comparison study. Enhanced GFP driven by a universal promoter was packaged into AAV9, AAV-PHP.eB, or PM-AAV9 and delivered by bilateral ICV injection into wildtype mice at P1. B Comparison of cortical transduction achieved by each capsid 3 weeks post-injection, with images taken at the same exposure for transduction comparison. C PM-AAV9 was re-imaged at lower exposure for more specific visualization of transduction including Layer V cortical cells. D Comparison of lumbar spine transduction by each capsid, with 10X image showing anterior horn cell transduction. Scale bars for (B) and (D) represent 250 μm. E Schematic of vector used in therapeutic studies, consisting of miAtxn2 driven by the mU6 promoter and packaged in PM-AAV9, herein referred to as miAtxn2. F ATXN2 protein levels in wildtype, TAR4/4, and miAtxn2-treated TAR4/4 mouse lumbar spine 3 weeks after treatment with miAtxn2, visualized by ATXN2 (green) and ChAT (red) immunofluorescence. Scale bar represents 250 μm. G Quantification of ATXN2 in ChAT+ anterior horn cells, normalized to untreated TAR4/4 mice. Values for individual mice are shown along with mean and standard error for each group. N = 5 mice/group for Wildtype and miAtxn2-treated mutant and 4 mice/group for Mutant, with 2-10 images analyzed per mouse. Data were analyzed by ordinary one-way ANOVA followed by Holm-Sidak post-hoc. *p < 0.05, ****p < 0.0001. Specific p-values: Wildtype vs Mutant, 0.0137; Mutant vs miAtxn2-treated, 0.0137; Wildtype vs miAtxn2-treated, <0.0001. Source data are provided as a Source Data file. Schematics in (A) and (E) were created in BioRender: Amado, D. (2025), [73]https://BioRender.com/r71o037 and Amado, D. (2025) [74]https://BioRender.com/n53n743, respectively. MiAtxn2 was packaged in PM-AAV9 and injected into the lateral ventricles at P1 for all further studies (Fig. [75]1E). In C57Bl/6/SJL wildtype mice, Atxn2 mRNA levels were measured 3 weeks after injection in ALS-relevant regions of the central nervous system (Fig. S[76]1Ci), demonstrating 54% knockdown in the frontal cortex (Fig. S[77]1Cii) and 23-25% knockdown in the brainstem (Fig. S[78]1Ciii), cervical spine (Fig. S[79]1Civ), and lumbar spine (Fig. S[80]1Cv) in mice treated with PM-AAV9.miAtxn2 (herein referred to as miAtxn2) vs mice treated with a non-Atxn2-targeting miRNA (miCtrl). Cortical ATXN2 protein levels were correspondingly reduced 32% in homogenized tissue by western blot (Fig. [81]S1D). There was no significant astrogliosis (Fig. [82]S1E) or microgliosis (Fig. [83]S1F) in response to treatment. In a cohort followed to 12 weeks, no dorsal root ganglia pathology was observed (Figure S[84]2A), with no morphologic differences or inflammatory infiltrates seen in the soma (Fig. [85]S2B) or nerve roots (Fig. [86]S2C) and no change in sensory neuron cell body size (Fig. [87]S2D, E). Effect of ATXN2 knockdown on survival, weight, and strength in TAR4/4 mice The efficacy of miAtxn2 was assessed in the TAR4/4 mouse model of sporadic ALS^[88]10,[89]12, a model with wildtype human TDP-43 pan-neuronally expressed starting at P7. Homozygous TAR4/4 mice develop an aggressive course of progressive weakness and weight loss, reaching endpoint in the fourth week of life. As disease advances, they lose motor neurons and display phosphorylated TDP-43 inclusions. Because of the rapid disease course and the delayed peak of AAV-mediated gene expression, TAR4/4 mice were treated at P1 by bilateral ICV injection (Fig. [90]1A). Rotarod performance and weight were measured at P17, and gait-related behaviors at P18, when disease is advanced but not end-stage. To mirror pre-IND-enabling studies, miAtxn2-treated mice were compared to buffer-treated mice. We first confirmed ATXN2 protein reduction in the lumbar spine of TAR4/4 mice. Western blot was felt to have low sensitivity for ATXN2 reduction in lower motor neurons, which are preferentially targeted by PM-AAV9 (Fig. [91]1D), as these cells constitute a small percentage of the total cellular constituents of the spinal cord. We therefore used immunohistochemistry on lumbar spine sections of TAR4/4 mice treated with miAtxn2 or buffer and compared these to wildtype levels (Fig. [92]1F). ATXN2 was lower in untreated TAR4/4 motor neurons than in those of untreated wildtype mice. Treatment with miAtxn2 resulted in a mean ATXN2 protein reduction of 65% in ChAT+ motor neurons with a maximum reduction of 100% (Fig. [93]1G). In TAR4/4 mice treated with miAtxn2, there was a 45.5% increase in median survival over buffer-treated mice (Fig. [94]2A). Mean survival was increased 54% (from 22 days ± 1 day to 33.5 days ± 3 days), with the longest-lived treated mouse surviving 58 days. A composite gait score was calculated for each mouse by scoring and summing parameters of abdominal droop, limping, foot angling, kyphosis, tremor, and clasping (Fig. [95]S3A). For the composite gait score (Fig. [96]2B) as well as for each of the sub-scores of limping (Fig. [97]S3B), foot angling (Fig. [98]S3C), kyphosis (Fig. [99]S3D), tremor (Fig. [100]S3E), and clasping (Fig. [101]S3F), the TAR4/4 phenotype was improved by miAtxn2 treatment. Rotarod duration, which was markedly decreased in TAR4/4 mice compared to wildtype, was improved with miAtxn2 treatment, with treated mice performing 2.6 times better than buffer-treated mice (Fig. [102]2C). Fig. 2. Effects of ATXN2 reduction on survival and strength in TAR4/4 mice. [103]Fig. 2 [104]Open in a new tab A Survival curve of miAtxn2-treated TAR4/4 mice compared to buffer-treated mice. N = 14 TAR4/4 and 15 miAtxn2-treated mice/group. Data were analyzed by both Log-rank [Mantel-Cox] test and Gehan-Breslow-Wilcoxon test. B Comparison of composite gait score at P18. N = 17 WT, 12 TAR4/4, and 14 miAtxn2-treated. C Comparison of endurance on accelerating rotarod at P17. N = 17 WT, 10 TAR4/4 (with 2 additional mice unable to perform test due to weakness), and 14 miAtxn2-treated. D Duration and frequency of vertical activity at P18. N = 17 WT, 11 TAR4/4, 12 miAtxn2-treated WT, 12 miAtxn2-treated TAR4/4. Values for individual mice are shown along with mean and standard error for each group. *p < 0.05, **p < 0.005, ***p < 0.0005, ****p < 0.0001, n.s. not significant. Data were analyzed by ordinary one-way ANOVA followed by Holm-Sidak post-hoc. WT, wildtype; MT, TAR4/4. Specific p-values: (A) 0.0003; (B) All comparisons <0.0001; (C) WT vs MT, <0.0001; MT vs MT+miAtxn2, 0.0025; WT vs MT+miAtxn2, <0.0001; (D) Duration: WT vs MT, 0.9001; WT vs WT+miAtxn2, 0.8091; MT vs MT+miAtxn2, 0.0051; WT+miAtxn2 vs MT+miAtxn2, 0.0016. Frequency: WT vs MT, 0.5283; WT vs WT+miAtxn2, 0.5267; MT vs MT+miAtxn2, <0.0001; WT+miAtxn2 vs MT+miAtxn2, <0.0001. Source data are provided as a Source Data file. The duration and frequency of vertical activity (Fig. [105]2D), which consists of both climbing and rearing behaviors and is considered a measure of hindlimb strength, were also assessed. While a TAR4/4 phenotype was not observed, there was an unexpected increase in vertical activity specifically in miAtxn2-treated TAR4/4 mice over wildtype levels, both in duration (2.4-fold over wildtype) and frequency (2.2-fold over wildtype). To assess the genotype specificity of this effect, we measured vertical activity in age-matched wildtype littermates. We found that there was no change in vertical activity frequency or duration in miAtxn2-treated wildtype mice, suggesting that this effect is specific to the TDP-43 overexpressing model. Effect of ATXN2 knockdown on cellular phenotypes Tissues from a subset of mice at P21-P23 were analyzed histologically. There was widespread apoptosis throughout the lumbar spine, as measured by the apoptotic marker cleaved caspase 3 (CC3); this was improved by treatment with miAtxn2 and fully corrected at the L3 and L4 levels (Fig. [106]3A, B). In the posterior lumbar spine there was loss of lower motor neurons, identified by ChAT positivity, that was rescued in miAtxn2-treated mice (Fig. [107]3C, D). Fig. 3. Lower motor neuron survival in TAR4/4 mice after miAtxn2 treatment. [108]Fig. 3 [109]Open in a new tab A Lumbar spinal sections from wildtype, TAR4/4, and miAtxn2-treated mice were stained with the apoptotic marker cleaved caspase 3 (CC3). Scale bar represents 250 μm. B CC3-positive cells were quantified at each of lumbar levels L3-L6 (L3 shown in A). N = 3–7 mice/group at each level (specifics below), 2–7 sections/mouse per level. C Lower motor neurons were identified by ChAT staining. Scale bar represents 250 μm. D ChAT-positive cells were quantified at each of lumbar levels L3-L6 (L6 shown in C). N = 4–6 mice/group at each level (specifics below), 4–10 sections/mouse per level. For (B) and (D), values for individual mice are shown along with mean and standard error for each group. Values from TAR4/4 mice treated with buffer or with miAtxn2 were normalized to wildtype levels from the same batch. *p < 0.05, **p < 0.005, ***p < 0.0005, ****p < 0.0001, n.s. not significant. Data were analyzed by two-way ANOVA split by mouse followed by Holm-Sidak post-hoc. LMN lower motor neuron; Mutant, TAR4/4. Specific N, listed in order of Wildtype, Mutant, and miAtxn2-treated: (B) L3: 6, 5, 3; L4: 7, 4, 4; L5: 4, 6, 3; L6: 5, 5, 4; (D) L3: 4, 3, 4; L4: 4, 3, 4; L5: 5, 3, 4; L6: 4, 3, 4. Specific p-values, listed in order of Wildtype vs Mutant, Mutant vs miAtxn2-treated, and Wildtype vs miAtxn2-treated: (B) L3: 0.0014, 0.0462, 0.1697; L4: <0.0001, 0.0001, 0.2163; L5: 0.0742, 0.9990, 0.0466; L6: 0.0808, 0.1263, 0.0316; (D) L3: 0.8722, 0.7787, 0.7366; L4: 0.0697, 0.7455, 0.0532; L5: 0.8611, 0.8611, 0.8611; L6: 0.0397, 0.0482, 0.3224. Source data are provided as a Source Data file. TAR4/4 mice showed a band of astroglial and microglial inflammation throughout cortical layer 5 that was reduced with miAtxn2 treatment (Fig. [110]4A–D), although there was no impact on Layer V neuron counts (Fig. [111]S3G). In the lumbar spine, we focused on spinal level L6, where there was motor neuron loss in TAR4/4 mice, to assess whether this was accompanied by gliosis. A similar gliosis phenotype to that seen in Layer 5 of the cortex was seen (Fig. [112]4E, H), with significant improvement in microgliosis following miAtxn2 treatment (Fig. [113]4F, H). Fig. 4. Effects of miAtxn2 on brain and spinal cord inflammation in TAR4/4 mice. [114]Fig. 4 [115]Open in a new tab Markers of neuroinflammation were stained and quantified in miAtxn2-treated and buffer-treated TAR4/4 mice and buffer-treated wildtype littermates. A, C Representative images of motor cortex at 5X (top row) magnification, with arrows indicating the gliosis in Layer 5 depicted below at 40X (bottom) magnification, stained for the astrocyte marker GFAP (A) or the microglial marker Iba1 (C). Scale bars represent 500 μm and 50 μm, respectively. B, D Quantification of the percent area of Layer 5 staining positive for GFAP (B) and Iba1 (D). For (B), N = 3 WT, 4 MT, 5 miAtxn2-treated, while for (D), N = 4 WT, 3 MT, 3 miAtxn2-treated; 3-6 sections/mouse. E, G Representative images of anterior horn of lumbar spine at 10X (top row) and 20X (bottom row) magnification, stained for the astrocyte marker GFAP (E) and the microglial marker Iba1 (G). Scale bars represent 250 μm and 100 μm, respectively. F, H Quantification of the percent area of the anterior horn staining positive for GFAP (F) and Iba1 (H). For (F), N = 7 WT, 6 MT, 5 miAtxn2-treated, while for (H), N = 6 WT, 5 MT, 6 miAtxn2-treated; 1-4 sections/mouse. For all graphs, values for individual mice are shown along with mean and standard error for each group, with data normalized to WT. *p < 0.05, **p < 0.01, ***p < 0.0005, n.s. not significant. Data were analyzed by ordinary one-way ANOVA followed by Holm-Sidak post-hoc. WT, Wildtype; MT or Mutant, TAR4/4. Specific p-values, listed in order of WT vs MT, MT vs miAtxn2-treated, and WT vs miAtxn2-treated: (B) 0.0071, 0.0211, 0.1887; (D) 0.0015, 0.0142, 0.0739; (F) 0.0005, 0.0776, 0.0275; (H) 0.0003, 0.0308, 0.0135. Source data are provided as a Source Data file. Pathologic TDP-43 is phosphorylated in TDP-43 protein-opathies^[116]13. TAR4/4 mice showed a strong increase in pTDP staining that normalized to WT levels after miAtxn2 treatment (Fig. [117]5A, B). This signal was prominent in motor neurons when lumbar spine sections were co-stained for pTDP, ChAT, and DAPI (Fig. [118]5C). To ensure this was not due to changes in total TDP-43 expression, we analyzed tissue from treated wildtype and hemizygous littermates of study mice, and found that in both the frontal cortex (Fig. [119]5D) and lumbar spine (Fig. [120]5E), total TDP-43 levels were not affected by treatment with miAtxn2. Fig. 5. Effects of miAtxn2 on TDP-43 levels and pathology. [121]Fig. 5 [122]Open in a new tab A Representative staining of phosphorylated TDP-43 (pTDP) in miAtxn2- or buffer-treated TAR4/4 mice and buffer-treated wildtype littermates at the L6 lumbar level. B Quantification of pTDP staining in the L6 anterior horn. N = 4 mice/group for WT and MT and 5 mice/group for miAtxn2-treated, 1-5 slides/mouse. Values for individual mice are shown along with mean and standard error for each group, with data normalized to WT. **p < 0.01, n.s. not significant. Data were analyzed by ordinary one-way ANOVA followed by Holm-Sidak post-hoc. Specific p-values: WT vs MT, 0.0043; MT vs miAtxn2-treated, 0.0068; WT vs miAtxn2-treated, 0.4572. C Confocal imaging of anterior horn cells stained with DAPI (blue), ChAT (red), and pTDP (green) at 40X magnification, with overlay shown in right panels; representative images from N = 3 mice/group are shown. Scale bars represent 50 μm. Total TDP-43 levels in treated and untreated wildtype and hemizygous TAR4 mouse frontal cortex (D) and lumbar spine (E) as measured by western blot analysis. N = 4 mice/group except for miAtxn2-treated WT for which N = 3. Values for individual mice are shown along with mean and standard error for each group, with data normalized to WT. *p < 0.05, n.s. not significant. Data were analyzed by unpaired two-tailed T-tests with comparisons shown on graph; a separate analysis by ordinary one-way ANOVA followed by Holm-Sidak post-hoc showed no significant differences between any comparison groups. WT Wildtype, MT or Mutant, TAR4/4; Hemi, TAR4 hemizygous. Specific p-values: (D) WT vs WT+miAtxn2, 0.6703; Hemi vs Hemi+miAtxn2, 0.2678; WT vs Hemi, 0.0116; (E) WT vs WT+miAtxn2, 0.3149; Hemi vs Hemi+miAtxn2, 0.7654; WT vs Hemi, 0.2918. Source data including uncropped blots are provided as a Source Data file. Effect of ATXN2 knockdown on transcriptional phenotypes To determine the disease-associated transcriptional signature of TAR4/4 mice, we dissected the lumbar spine when there is pronounced disease at P19 (Fig. [123]6A) and determined differential gene expression (DGE) between mutant and wildtype mice using bulk-RNA sequencing. Six WT, five TAR4/4, and five miAtxn2-treated mice were included in analysis, balanced across litters such that each litter contained at least one mouse in each group. In total, 1288 genes were dysregulated as a result of TDP-43 overexpression (adjusted p-value < 0.05; | log[2](fold change) | > 0.5); Figs. [124]6B, [125]S4A). Gene enrichment analysis identified amyotrophic lateral sclerosis as the pathway term with the greatest gene overlap, as well as other processes known to be dysregulated in ALS such as calcium signaling, apoptosis, and steroid biosynthesis (Figs. [126]6C, [127]S4B). Fig. 6. miAtxn2 treatment effect on transcriptomic dysregulation in TAR4/4 mice. [128]Fig. 6 [129]Open in a new tab A Schematic of experimental design for bulk transcriptomics of gene rescue in the lumbar spine of miAtxn2-treated TAR4/4 mice. B Volcano plot of differential gene expression between buffer-treated TAR4/4 (mutant) and buffer-treated wildtype (wildtype) mice. Significant differentially expressed genes are denoted in red (DESeq2 two-sided Wald test with Benjamini-Hochberg post-hoc, adjusted p-value < 0.05; | log[2](fold change) | > 0.5). C Lollipop plot of the top KEGG pathways enriched in genes dysregulated in mutant mice, identified in (A). richFactor and log[10](adjusted p-value) were calculated with the enrichR package using Fisher’s exact test. D Venn diagram of DEGs between mutant and wildtype mice and miAtxn2 and wildtype mice. 412 genes were identified as “normalized” or no longer meeting our criteria for differential expression (DESeq2 two-sided Wald test with Benjamini-Hochberg post-hoc, adjusted p-value > 0.05; |log[2](fold change) | <0.5) between mutant and wildtype mice upon miAtxn2 treatment. E Volcano plot of the log[2](fold change) and adjusted p-value for mutant vs. wildtype DEGs identified in (A) in response to miAtxn2 treatment. Blue denotes genes that were previously significantly expressed, but no longer meet these criteria upon miAtxn2 treatment, as shown by the Venn diagram in (D). F Lollipop plot of the top KEGG pathways enriched in genes normalized upon miAtxn2 treatment, identified in (E). richFactor and log[10](adjusted p-value) were calculated with the enrichR package, which uses Benjamini-Hochberg post-hoc. G Tukey box plots showing the gene expression of selected top gene hits between mutant vs. wildtype and miAtxn2 vs. wildtype, across all experimental groups. H Tukey box plots of selected top hits from miAtxn2 vs. mutant mice. I Tukey box plots of Tardbp gene expression across experimental groups. For each boxplot in (G–I), each individual point corresponds to a single mouse; the central line corresponds to the median (50th percentile); the lower and upper bounds correspond to the first and third quartiles (25th and 75th percentile) respectively; and the upper and lower whisker lines extend to the largest and smallest values within 1.5x interquartile range, respectively. Mutant, TAR4/4; DEG differentially expressed genes. Schematic in (A) was created in BioRender: Robbins, A. (2025), [130]https://BioRender.com/6t575wq. Of the ~1300 genes differentially expressed in TDP-43 mice relative to wildtype mice, 452 genes were corrected with miAtxn2 treatment (Figs. [131]6D, [132]S4C). To visually illustrate the treatment-induced shift of the TAR4/4 disease signature (Fig. [133]6B) towards wildtype levels, the results of the DGE analysis between miAtxn2 and wildtype littermates were plotted for the subset of 1288 genes (Figs. [134]6E, [135]S4D). Pathway enrichment analysis of the normalized genes demonstrated biosynthetic processes, tissue homeostasis, and cell proliferation (Fig. [136]6F), and among these, 48 had expression levels in miAtxn2-treated mice that were no longer statistically significant from wildtype (adjusted p-value > 0.05; Fig. [137]S4E). We note rescued expression of genes involved in extracellular matrix organization (e.g., Fbln5 and Itga10)^[138]14–[139]16, the TCA cycle (e.g., Idh3a)^[140]17, and DNA repair (e.g., Hist2h2aa1)^[141]18,[142]19, all of which were downregulated in TAR4/4 mice^[143]14–[144]16. We also note correction of signal transduction pathway genes that were up-regulated (e.g., Trpc6, Hif3a) or down-regulated (e.g., Vip, Vdr) in TAR4/4 compared to wildtype littermates^[145]20–[146]24 (Figs. [147]6G, [148]S4E). Additionally, 221 genes differentially expressed between miAtxn2-treated TAR4/4 mice and wildtype littermates were not in the original disease signature (Fig. [149]6D). To better assess the effect of miAtxn2 treatment, while controlling for genotype, we compared miAtxn2-treated mice to mutant littermates (Fig. [150]S4F). 93 differentially expressed genes (DEGs) were identified (adjusted p-value < 0.05; | log[2](fold change) | > 0). Gene ontology analysis of this miAtxn2 gene signature identified pathways involved in neurogenesis, myelination, axonogenesis and sphingolipid binding (Fig. [151]S4G). 23 of the 93 DEGs overlapped with our TAR4/4 disease signature, a literature review of which showed them to be implicated in ALS and/or neurodegenerative pathogenesis. For example, Dbr1, a gene previously identified as a potential therapeutic target for ALS^[152]24, was upregulated in TAR4/4 mice and normalized toward wildtype with miAtxn2 treatment (Fig. [153]6H; Table [154]S1). Similar to prior reports^[155]8 and corroborating our western blot analysis, we found an expected increase in total TDP-43 transcript levels in TAR4/4 mice that was not affected by Atxn2 reduction (Fig. [156]6I). Long-term effects of treatment in hemizygous and wildtype mice The hemizygous TAR4 mice in our colony show clasping onset at 3 months with emerging gait dysfunction shortly thereafter (Fig. [157]7A), which is an earlier onset than previously reported^[158]10. Gait dysfunction steadily progressed in buffer-treated TAR4 mice, but did not progress from 3 to 12 months in miAtxn2-treated TAR4 mice after which it progressed slowly. miAtxn2 treatment of wildtype littermates resulted in no phenotypic changes over 21 months of observation (Fig. [159]7B). Fig. 7. Effect of miAtxn2 on gait function in TAR4 hemizygous and wildtype mice. [160]Fig. 7 [161]Open in a new tab TAR4 hemizygous mice and their wildtype littermates were treated with miAtxn2 or buffer at P1 and composite gait score was measured at 3 month intervals. A Effect of miAtxn2 on TAR4 gait over time. B Effect of miAtxn2 on WT gait over time. N = 11–21 mice/group at 3–6 months, 18–37 mice/group at 9–18 months, and 9–17 mice/group at 21 months (specifics below). Values for individual mice are shown along with mean and standard error for each group. *p < 0.05, **p < 0.01, ***p < 0.005, ****p < 0.001, n.s. not significant. Data were analyzed by two-way ANOVA followed by Holm-Sidak post-hoc. WT wildtype, Hemi, TAR4 hemizygous. Specific N: For (A), listed in order of Wildtype, Mutant, and miAtxn2-treated mutant: 3 months: 11, 21, 21; 6 months: 11, 20, 21; 9 months: 19, 19, 31; 12 months: 18, 35, 24; 15 months: 23, 37, 33; 18 months: 17, 31, 23; 21 months: 9, 9, 17. For (B), listed in order of Wildtype and miAtxn2-treated wildtype: 3 months: 11, 14; 6 months: 11, 9; 9 months: 19, 14; 12 months: 18, 12; 15 months: 23, 15; 18 months: 17, 9; 21 months: 9, 6. Specific p-values: For (A), listed in order of Wildtype vs Mutant, Mutant vs miAtxn2-treated, and Wildtype vs miAtxn2-treated: 3 months, 0.0008, 0.4374, 0.0065; 6 months, <0.0001, 0.2464, <0.0001; 9 months, <0.0001, 0.7395, <0.0001; 12 months, <0.0001, 0.0184, 0.0008; 15 months, <0.0001, 0.0102, <0.0001; 18 months, <0.0001, 0.1635, <0.0001; 21 months, 0.0030, 0.2302, 0.0030. For (B), none significant (range: 0.7264-0.9935). Source data are provided as a Source Data file. Translatability to human targets To determine the ability of miAtxn2 to target human ATXN2, we used the 127Q transgenic mouse model of spinocerebellar ataxia type 2, in which mice overexpress mutant human ATXN2 in cerebellar Purkinje cells. After confirming targeting of affected cells by PM-AAV9.eGFP after ICV delivery at P1 (Fig. [162]8A), PM-AAV9.miAtxn2 delivery potently reduced human ATXN2 levels in the cerebellum (Fig. [163]8B). Fig. 8. miAtxn2 targeting of human ATXN2 and transduction of ALS patient-derived lower motor neurons. [164]Fig. 8 [165]Open in a new tab A Mouse cerebellar Purkinje cell transduction after delivery of PM-AAV9.eGFP, representative image from N = 4 mice. Scale bar represents 250 μm. B Reduction of human ATXN2 mRNA in cerebellum of 127Q transgenic mice, which express ATXN2 exclusively in Purkinje cells, after treatment with miAtxn2 compared to a non-ATXN2-targeting control (miCtrl). N = 5 mice/group. Values for individual mice are shown along with mean and standard error for each group. ****p < 0.0001. Data were analyzed by unpaired two-tailed T-test. C Confirmation of lower motor neuron identity of a differentiated C9orf72-ALS patient cell line using NeuN and ChAT markers; representative images from N = 3 replicates per marker are shown. Scale bar represents 100um. D Representative image and quantification of GFP positivity in patient-derived lower motor neurons after transduction with PM-AAV.miAtxn2. Scale bar represents 250 μm. N = 3 replicates per dose, and mean and standard error are shown. ****p < 0.0001. Data were analyzed by two-way ANOVA followed by Holm-Sidak post-hoc. E Dose-dependent reduction in ATXN2 levels in patient-derived lower motor neurons, without affecting total TDP-43 transcript levels (F), N = 2 replicates per experiment. MOI, multiplicity of infection. For (D–F), each dot represents the averaged value of five images from an individual well. Source data are provided as a Source Data file. We next assessed the ability of PM-AAV9 to transduce human lower motor neurons, using a cell line derived from a C9orf72-ALS patient with 800 repeats that was engineered to contain an integrated doxycycline (DOX)-inducible transposon expressing lower motor neuron-differentiating factors^[166]25. Upon DOX induction, lower motor neuron identity was confirmed by NeuN and ChAT staining (Fig. [167]8C). Prior studies using AAV have required MOI of up to 2 × 10^8 to transduce patient-derived lower motor neurons^[168]26. PM-AAV9 was used to deliver GFP and resulted in 70% neuronal transduction at MOI of 5 × 10^4 that increased to >90% at MOI of 5 × 10^5 (Fig. [169]8C, D). PM-AAV9.miAtxn2 delivery resulted in dose-dependent ATXN2 mRNA reduction (Fig. [170]8E), achieving 70% reduction and 83% reduction at MOI of 5 × 10^4 and 5 × 10^5, respectively, without affecting total TDP-43 transcript levels (Fig. [171]8F). Discussion In this study, we developed an RNAi-based method to reduce ATXN2 in a mouse model of sporadic ALS. We achieved potent reduction of target transcripts in ALS-relevant regions including the motor cortex, brainstem, and spinal cord without impacting total TDP-43 transcript levels, which in turn improved motor function and survival. Interestingly, we also observed increased vertical activity above wildtype levels. While Atxn2-null mice have locomotor hyperactivity, in our study only the TAR4/4 mice (and not their wildtype littermates) showed this effect of ATXN2 reduction, which therefore may represent an unmasking of impulsivity (i.e., an FTD-like phenotype) in the setting of improved motor function. Histopathologically, we observed markedly reduced inflammation in cortex and spinal cord, reduced apoptosis in proximal levels of the lumbar spine, and distal-predominant rescue of lower motor neurons with normalization of phosphorylated TDP-43 levels. This was accompanied by improvement in many genes dysregulated across the transcriptome, including several described in human ALS literature (see “Supplementary Information” bibliography). In the slower-progressing hemizygous littermates, there was significant slowing of progression in gait dysfunction with no adverse effect on gait in long-term study of wildtype littermates. The potential for translatability to human studies was shown by highly efficient transduction and ATXN2 reduction in lower motor neurons at MOIs nearly 4 logs lower than published literature. These findings corroborate and elaborate on the functional and survival benefits seen in this model with ASO-based ATXN2 knockdown^[172]8, as well as findings described using a CRISPR/Cas13 system^[173]27. Notably, a recent ATXN2-targeting ASO was tested in human subjects with either sporadic ALS or ALS caused by trinucleotide repeat expansions in the ATXN2 gene. While the trial did not meet its target endpoints for either population of patients, only 21% reduction of ATXN2 protein from baseline levels was seen in the CSF and only a 12% reduction when compared to placebo (in whom ATXN2 levels also declined from baseline), a level that fell far short of the expected ~80% based on preclinical studies. We are not convinced that this is a sufficient degree of knockdown to expect a therapeutic effect, particularly given that unlike Tofersen for SOD1-ALS, the trial drug targets an indirect modifier of pathology in sporadic patients. The 12% ATXN2 reduction corresponded to the maximal safe dose of ASO, leading to complete cessation of the trial as the dose could not be increased. This highlights the need for therapies that can achieve strong target engagement in affected cells at safe doses. Modified AAV vectors like the one used in this study offer a means of improving CNS transduction of disease-relevant regions and cell types at relatively low therapeutic doses. From a translational standpoint, there are additional advantages of our approach. The first is long-term benefit after a single treatment, obviating the need for repeated CNS access. In the ALS patient population, physical limitations and the need for access to a tertiary care center are limiting factors in the broad application of ASO-based therapies that are overcome by AAV-based treatments. The second is that the PM-AAV9 doses used here achieved efficacy at ~1/40th the dosage of AAV9 used in the Cas13 mouse study, and at ~1/3600th the dosage of AAV5 used in an AAV-mediated RNAi study in patient-derived lower motor neurons. This is important in translating these therapies by reducing both production costs and known AAV complications such as dose-dependent dorsal root ganglia (DRG) toxicity^[174]28. We did not see evidence of DRG toxicity in our study at 12 weeks either in numbers of DRG neurons or presence of inflammatory cells in the DRGs (Fig. [175]S2), nor did we see DRG-associated clasping phenotypes^[176]28 in PM-AAV9-treated wildtype mice even as late as 21 months (Fig. [177]6). The third is that RNAi-based approaches have a strong safety and efficacy record in humans, with lipid nanoparticle-based delivery in current clinical use (e.g., patisiran for TTR amyloidosis) and AAV-based RNAi approaches in clinical trial (e.g., [178]NCT06100276, AAV-RNAi for SOD1-ALS; [179]NCT04120493, AAV-RNAi for Huntington’s disease, with recently reported positive Phase II data^[180]29). By contrast, CRISPR-based treatments have been used safely in humans only for transient delivery systems, and at this time RNAi may be more amenable to AAV-based expression from both an off-targeting and immunogenicity standpoint with permanently-on expression. From a further safety standpoint, a recent study demonstrated long-term safety of the miR30 miRNA cassette^[181]30, a modified version of which was used in our work^[182]31. A limitation of our approach and the prior Atxn2 knockdown studies^[183]8,[184]27 is that all three were tested in the TAR4/4 mouse model. This mouse overexpresses wildtype human TDP-43, and was chosen because it is one of few models that replicate important human ALS findings at the pathologic and behavior level, including pTDP aggregates and progressive weakness respectively. Our study also provides the first glimpse at therapeutic benefit in the slower-progressing hemizygous mice, which overexpress half as much TDP-43 as the TAR4/4 mice. However, because patient neurons do not overexpress TDP-43 but rather mislocalize it, it remains unknown whether the Atxn2 knockdown benefit seen in our and other studies is unique to TDP-43 overexpression. An additional limitation is that TAR4/4 mice model ALS-relevant toxic gain-of-function but not loss of function^[185]5,[186]6. The fact that rescue is noted suggests that addressing toxic gain-of-function may be sufficient for therapeutic effect in patients, but further studies are needed to determine if there is efficacy of Atxn2 knockdown in non-TDP-overexpressing model systems. Transcriptomic analysis revealed several mechanisms by which ATXN2 reduction may be exerting neuroprotection. Lipid dysmetabolism is a well-documented feature of ALS pathology^[187]32–[188]34, with elevated levels of sphingolipids in the CSF and plasma of ALS patients negatively correlating with prognosis^[189]35,[190]36. Atxn2 knockout mice present with dyslipidemia^[191]37—which has been suggested to be protective in ALS^[192]38—and specifically with reduced levels of sphingomyelin and intermediate metabolites^[193]37,[194]39. Our miAtxn2-responsive TAR4/4 gene signature was enriched in fatty acid and sphingolipid biosynthetic pathways^[195]39, suggesting a role of ATXN2 in the regulation of sphingomyelin metabolism and a potential mechanism by which its reduction may be neuroprotective. ATXN2 reduction also normalized neuroinflammatory pathway transcripts, including via the upregulation of vascular endothelial growth factor A (Vegfa) and its modulator vasoactive intestinal peptide (Vip) towards wildtype levels. These are both known regulators of neuroinflammatory processes^[196]40,[197]41 whose protein levels are reduced in the cerebrospinal fluid of ALS patients^[198]22,[199]42, and multiple studies have demonstrated their therapeutic potential through rescue of ALS-associated gliosis and prolonged survival in rodent models^[200]43–[201]45. This mirrors the rescue of gliosis in the cortex and lumbar spine demonstrated by immunohistochemistry. We also note normalization of genes involved in oxidative stress, including the PI3K-Akt signaling pathway^[202]46 (Fig. [203]S3B), and rescue of immediate early gene transcription factor early growth response 3 (Egr3) expression, which is also downregulated in motor neurons in sporadic ALS^[204]47,[205]48 and is a key regulator of neuronal activity-dependent transcription and DNA damage response genes pathways^[206]49,[207]50. Finally, miAtxn2 treatment rescued TDP-43 induced overexpression of RNA lariat debranching enzyme Dbr1, whose reduction was recently shown to modify TDP-43 toxicity by decreasing TDP-43 cytoplasmic aggregation and increasing neuronal viability^[208]24. Future transcriptomic studies in other model systems and in TAR4 hemizygous mice will help determine the extent to which these results are generalizable to human disease and whether they are dependent on the degree of TDP pathology, respectively. An important limitation of this analysis is that bulk-RNA sequencing includes all cell types in the tissue, of which motor neurons comprise a small percentage. This approach enables analysis of global changes across cell types but precludes inference about which changes are specific to motor neurons. Therefore, future studies using single-cell or spatial transcriptomics approaches will be of benefit for parsing cell-autonomous changes. In sum, we developed an RNAi-mediated ATXN2 knockdown approach that shows benefit in rapid and slow progressing TDP-43 mouse models of ALS, which capture a range of typical human disease time-courses. We further identified potential mechanisms by which ATXN2 reduction leads to neuroprotection and demonstrated translatability in human motor neurons. Vectors with increased potency and specificity have the potential in humans to achieve the target engagement that ASOs did not, and further may reduce AAV dosage and off-targets, which in combination with RNAi can provide a translatable therapeutic approach for people living with sporadic ALS. Methods miRNA development and validation Artificial miRNAs were generated by polymerase extension of overlapping DNA oligonucleotides (IDT, Newark, NJ), purified using Qiaquick PCR purification kit, digested with XhoI/SpeI, and cloned into a XhoI/XbaI site in a miR30-like modified Pol-III expression cassette containing the mouse U6 promoter, a multiple cloning site, and the Pol-III terminator (6Ts)^[209]31. miRNA sequences were designed using siSPOTR, an algorithm developed to identify potent RNAi sequences with low unintended off-target profiles^[210]51. Additionally, the top RNAi sequence was further engineered for optimal strand loading of the guide strand and for reduced profile silencing of the passenger strand. miRNAs were screened for Atxn2 knockdown by transfection into mouse neuroblast (N2A) cell lines. For in vivo studies, the miRNA expression cassettes were cloned into an AAV shuttle plasmid upstream of a DNA stuffer sequence. The stuffer sequence was obtained by amplification and assembly of intronic sequences of human HTT and was designed to be devoid of enhancer or repressor sequences, splice activators or repressors, and antisense or other non-coding RNAs^[211]52. The miRNA expression cassette and stuffer sequence were flanked at each end by AAV2 145-bp inverted terminal repeat (ITR) sequences. AAV vector selection AAV9, AAV-PHP.eB, and a novel peptide-modified AAV9 capsid variant, PM-AAV9, developed in our lab^[212]11 were each used to package eGFP under the universal CMV immediate enhancer/beta-actin (CAG) promoter, with vectors prepared by the Children’s Hospital of Philadelphia Research Vector Core. These were delivered through bilateral intracerebroventricular (ICV) injection into the brain of P1 mice at a dose of 2E10 VG/pup in 4uL total. Brains and spinal cords were harvested and sectioned 3 weeks post-injection and imaged on a Leica (Wetzlar, Germany) DM6000B fluorescence microscope for native fluorescence using LAS X software. The top performing capsid, PM-AAV9, was used to package the top-performing miRNA variant to generate the therapeutic used throughout subsequent studies. A non-targeting control miRNA, miCtrl, was prepared as previously described^[213]53 and was similarly packaged and used for comparison studies of knockdown efficacy in vivo. Animals This study was conducted in accordance with the Guide for the Care and Use of Laboratory Animals (National Research Council), and all procedures were approved by the Children’s Hospital of Philadelphia Animal Care and Use Committee under protocol number 23-001358. Mice were housed at the CHOP Research Institute in a controlled temperature and humidity environment on a 12 h light/dark cycle. Food and water were provided ad libitum. TDP-43 transgenic mice generated by Samir Kumar-Singh (TAR4 strain) were purchased from JAX (strain 012836, B6;SJL-Tg(Thy1-TARDBP)4Singh/J). Mice were maintained on a B6/SJL background by crossing hemizygous mice with F1 hybrid mice from JAX (strain 100012, B6SJLF1/J). Hemizygous mice were crossed with one another to generate the TAR4/4, TAR4, and WT offspring used in our studies. Male and female mice were used for all studies based on published reports of a lack of sex-based phenotypic differences in this model^[214]8,[215]10. Pups were weaned at P21-P23. All litters were provided with DietGel Boost High Calorie Dietary Supplement (ClearH[2]O, Westbrook, ME) starting at P12 that was placed on the floor of the cage for easy access by impaired mice. Genotyping Mice were genotyped at P6 via tail snip, using the common TDP primer 5’-TGAAATCCGGGTGGTATTGG-3’ (13790, JAX, Bar Harbor, ME), wildtype TDP primer 5’-GGTGAGTTTAACCTTCAAGGGCT-3’ (13791, JAX), and transgene TDP primer 5’-AGCTTGCTAGCGGATCCAGAC-3’ (13792, JAX). Prior to weaning, mice were individually identified by skin-safe colored markers (Stoelting, Wood Dale, IL) and by ear-snips after weaning. Injections Pups were anesthetized at P1 by hypothermia and their heads disinfected with 70% ethanol, and injections performed on a cold plate. Mice were injected in the bilateral cerebral ventricles with a total of 2E10 VG/pup (2uL per hemisphere of 5E12 VG/mL), at coordinates 1/3 the distance between the lambda suture and the eyeball (~2 mm anterior to lambda and 1 mm lateral to midline) and at a depth of 2 mm, using a 33-gauge 10 uL Hamilton syringe (Hamilton Company, Reno, NV). After each injection the needle was held in place for ten seconds prior to withdrawal. Mice were re-warmed prior to returning them to their cage. Mice were treated by litter to avoid administering tattoos so early in life, out of concern that related complications (e.g., paw damage) could interfere with downstream behavior assays. Vectors were prepared by a third party, and the person administering the injection was blinded to treatment. Behavioral testing Behavioral tests were conducted by an investigator blinded to the genotype and treatment of mice. All testing was performed during the light period. Mice were habituated to the test room for 1 h and were given Clear H[2]O HydroGel if testing lasted more than 4 h. Work surfaces and equipment were cleaned with 70% ethanol between trials and then wiped with water to remove the ethanol scent. N = 10–40 mice/group for TAR4/4 studies in Fig. [216]2, and 11–31 mice/group for the TAR4 studies in Fig. [217]7. Rotarod analysis Mice were tested on accelerating rotarod (47600; Ugo Basile, Gemonio, Italy) at P17. Mice were trained for a single trial at a constant speed of 2 rpm for 5 min. Mice were tested for 3 trials with a starting speed of 2 rpm and acceleration to 20 rpm over 4 min, followed by a hold at 20 rpm for 1 min. Latency to fall (or two consecutive rotations without running) was recorded for each trial, and mice were given 30 min of rest between trials. Mice were weighed at the end of the session. Gait analysis Treated and untreated TAR4/4 mice and untreated WT littermates underwent gait testing on P18. Additionally, treated and untreated TAR4 mice and treated and untreated WT littermates underwent gait testing every 3 months for 21 months. Mice were recorded using a Panasonic HD Camcorder HC-V180. Gait videos were analyzed for composite gait score (adapted from ref.^[218]54) consisting of hindlimb clasping, abdomen height, limping, foot angling, kyphosis, and tremor. Composite gait was analyzed by 2 independent scorers blinded to treatment and genotype. Hindlimb position was recorded for 30 s with mice held 1 cm from the tip of the tail. Mice received a score of 0 if the hindlimbs were consistently splayed outward. If a single hindlimb was retracted toward the abdomen for more than 50% of the time suspended, the mouse received a score of 1. If both hindlimbs were partially retracted for more than 50% of the time suspended, the mouse received a score of 2. If both hindlimbs were completely retracted for more than 50% of the time suspended, the mouse received a score of 3. If both hindlimbs were completely retracted for more than 90% of the time suspended or if the hindlimbs were both retracted with toes clasped together, the mouse received a score of 4. Gait was tested immediately following hindlimb clasping. Mice were recorded walking for 150 s on a flat plastic gait walkway. If a mouse did not move for longer than 20 s, the tester tapped them lightly on the back to encourage walking. Abdomen height was scored as a 0 if the abdomen was raised, or a 1 if the abdomen was lowered or touching the ground. Limping was scored as a 0 if the mouse was not limping or a 1 if the mouse exhibited a limp. Foot angling was scored as a 0 if the feet were aligned during walking or a 1 if the feet were pointed away from the body while walking. Mice received a kyphosis score of 0–3 based on the criteria outlined by Guyenet et al.^[219]54. Mice received a tremor score between 0 and 3, with 0, 1, 2, and 3 representing no, slight, moderate, or severe tremor, respectively. Composite gait score between 0 and 13 was calculated by adding individual scores for hindlimb clasping, abdomen height, limping, foot angling, kyphosis, and tremor. Vertical activity Mice underwent vertical activity testing following gait testing on P18, with at least 30 min of rest between tests. Mice were placed underneath wire mesh pencil holders (11 cm diam. x 17 cm height) and filmed for 5 min. Videos were scored for frequency and duration of rearing and climbing by an independent scorer who was blinded to genotype and treatment. Rearing was defined as the mouse leaning against the side of the cup on its forelimbs, and climbing was defined as all four limbs off the ground. Survival and humane endpoint determination All mice were observed daily for signs of weakness. Once severely impaired, they underwent daily righting trials consisting of laying them supine and allowing 30 s to right themselves, repeated up to three times. Euthanasia endpoint was defined as inability to right themselves in any of the 3 trials. Mice were also weighed 3 times a week throughout the study and were considered to have reached humane endpoint if they lost more than 25% of their maximum body weight. Examiners were blinded to genotype and treatment. Tissue collection and preparation Mice were euthanized at P21 for RNA analyses and vector distribution studies, and at P22-24 for immunofluorescence (IF) and immunohistochemistry (IHC) studies. Mice were deeply anesthetized with isoflurane and transcardially perfused with ice-cold phosphate-buffered saline (PBS), followed by 4% paraformaldehyde perfusion for immunofluorescence (IF) or immunohistochemistry (IHC) studies. Brain was dissected out, and spinal cord was removed by hydraulic extrusion, and both washed in chilled PBS. For RNA or protein analyses, this was followed by microdissection and flash-freezing in liquid nitrogen. For IF and IHC studies, tissues were immersed in 4% paraformaldehyde in PBS at 4 °C overnight, then transferred to a 30% sucrose solution in 0.05% azide/PBS for 48 h prior to embedding in Tissue-Tek O.C.T. compound and storing at −80 °C. RNA extraction and quantitative PCR RNA was extracted from relevant tissues or cells using Trizol (15596018; Thermo Fisher Scientific, Waltham, MA), with RNA quantity and quality measured using a NanoDrop 2000 (Thermo Fisher Scientific). Random-primer first-strand cDNA synthesis was performed using 1 μg of total RNA and a High Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Waltham, MA) per manufacturer’s instructions. Assays were performed on a Bio-Rad (Hercules, CA) CFX384 Real Time System using TaqMan (Thermo Fisher Scientific) primer/probe sets specific for mouse Actb, mouse Atxn2, mouse Gfap, or mouse Aif1 in the case of mouse tissue, and human ATXN2, human TARDBP, or human GAPDH in the case of iPSCs (Thermo Fisher Scientific FAM TaqMan 2X Universal Master Mix, Life Technologies, Waltham, MA). Protein extraction and western blot Frozen frontal cortex tissue samples were weighed and homogenized with a pestle in 10 μL/mg of extraction buffer. For ATXN2 quantification, RIPA buffer was used (50 mM Tris buffer [pH 8.0], 150 mM NaCl, 1% Triton X-100, 0.1% SDS, 0.5% sodium deoxycholate), and samples were incubated on ice for 30 min with vortexing every 10 min, then centrifuged at 16,000 x g for 20 min at 4 °C. Total protein concentration was quantified with the Peirce BCA Protein Assay kit (23225; ThermoScientific, Waltham, MA), and 30 μg of total protein was diluted with 7.5 μL of 4X Laemmli buffer (1610747; BioRad, Hercules, CA) in RIPA. For TDP-43 quantification, urea buffer was used (7 M urea, 2 M thiourea, 4% CHAPS, 50 mM tris buffer [pH 8.5]) and samples were incubated at RT for 20 min with vortexing every 5 min, then centrifuged at 21,100 x g for 30 min at 4 °C. 10uL of supernatant was diluted with 7.5 μL of 4X Laemmli buffer in 12.5uL of RIPA. Samples were run on 10–20% Criterion Tris-HCl gels (3450042; BioRad) and transferred to PVDF membranes. Membranes were blocked in 5% milk in TBS-T (0.1% Tween-20) at RT for 1.5 h, followed by incubation for 2 h at RT with primary antibodies in 5% milk in TBS-T at the following dilutions: anti-TDP-43 rabbit polyclonal antibody (1:1500; 10782-2-AP; Proteintech, Rosemont, IL), anti-Gapdh mouse monoclonal antibody (1:12,000; sc-32233; Santa Cruz Biotechnology, Dallas, TX), anti-Ataxin 2 rabbit polyclonal antibody (1:4000; 21776-1-AP; Proteintech), anti-Lamin B1 rabbit polyclonal antibody (1:4000; ab16048; Abcam, Cambridge, UK) or anti-beta Catenin rabbit polyclonal antibody (1:4000; ab2365; Abcam). Membranes were washed x3 with TBS-T for 5 min and treated with goat anti-mouse IgG (H + L) secondary antibody HRP (1:10,000; 31,430; Invitrogen, Waltham, MA) or goat anti-rabbit IgG (H + L) secondary antibody HRP (1:10,000; 31,460; Invitrogen) in 2% milk in TBS-T at 4 °C for 1.5 h. Membranes were washed x3 with TBS-T for 5 min and developed with Amersham ECL reagent (RPN2236, Cytiva, Marlborough, MA), and imaged on a BioRad ChemiDoc MP Imaging System. N = 3–4 mice/group, all from the same litter. Histology Embedded tissues were cut at 16μm thickness at −20 °C using a Leica CM 3050S Cryostat and stored at −80 °C. Primary antibodies used for tissue immunostaining included the following: anti-ATXN2 rabbit polyclonal antibody (1:500; 21776-1-AP; Proteintech); anti-TDP-43 phospho-Ser409/410 rabbit polyclonal antibodies (1:200; 22309-1-AP; Proteintech | 1:2000; 80007-1-RR; Proteintech); anti-ChAT goat polyclonal antibody (1:400; AB144P; Millipore, Burlington, MA); anti-NeuN rabbit monoclonal antibody (1:1000; EPR 12763; Abcam); anti-Gfap rabbit polyclonal antibody (1:2000 [brain] or 1:4000 [spinal cord]; Z0334; Dako/Agilent, Santa Clara, CA); anti-Iba1 rabbit polyclonal antibody (1:2000 [brain] or 1:4000 [spinal cord]; 019-19741; Wako, Richmond, VA); and anti-Cleaved Caspase-3 polyclonal antibody (1:400; 9661; Cell Signaling, Danvers, MA). All primary antibodies were diluted in 2% serum (of the secondary antibody species) in 0.1% Triton-X100/PBS. Secondary antibodies used for immunofluorescence included Donkey anti-rabbit AlexaFluor 488 (A-21206), Donkey anti-mouse Alexafluor 488 (A-21202), and Donkey anti-goat AlexaFluor 568 (A-11057) polyclonal IgG antibodies at a concentration of 1:1000 (ThermoScientific, Waltham, MA). Secondary antibodies used for immunohistochemistry included biotinylated goat anti-rabbit (BA-1000) and biotinylated horse anti-goat (BA-9500) at a concentration of 1:200 (Vector Laboratories, Burlingame, CA). Embedded dorsal root ganglia were cut at 12 μm thickness and washed in PBS to remove O.C.T. medium. Sections were stained with Harris’ Hematoxylin for 1 min (26041-05; Electron Microscopy Sciences), differentiated with 1% acid ethanol, blued in 1% sodium acetate, and counterstained with 0.5% Eosin Y (HT110216; Sigma-Aldrich, Millipore Sigma) for 1 min. Slides were cover-slipped using DPX (06522; Sigma-Aldrich). For immunofluorescence, slides were washed in PBS to remove O.C.T. medium and then blocked in 10% donkey serum in 0.1% Triton-X100/PBS at RT for 1 h. Sections were incubated in primary antibody (diluted in 2% donkey serum in 0.1% Triton-X100/PBS) overnight at 4 °C. Slides were washed in PBS and then incubated in secondary antibody (in 2% donkey serum in 0.1% Triton-X100/PBS). Slides were counterstained in Hoechst at 1:5,000 in PBS. Slides were coverslipped using Fluoromount-G (0100-01; SouthernBiotech, Birmingham, AL). For immunohistochemistry, slides were washed in PBS to remove OCT, then transferred to 5:1 methanol:30% hydrogen peroxide for 30 min to block endogenous peroxidase. Slides were triple-washed for 5 min at RT in dilution media (0.1% Tris-base, 0.0002% NaCl, and 0.2% Triton-X) and then blocked in 10% goat or donkey serum in 0.1% Triton-X100/PBS. Sections were then incubated in primary antibody (diluted in 2% serum in 0.1% Triton-X100/PBS) overnight at 4 °C in humidified chambers, PBS-washed, and incubated in similarly diluted secondary antibody for 1 h at RT. Sections were then treated for one hour at RT with the VECTASTAIN ABC kit (PK-6100; Vector laboratories) for avidin binding and peroxidation, then treated with Vector ImmPACT 3,3′-Diaminobenzidine (DAB) peroxidase substrate solution for detection (SK-4105; Vector laboratories). Slides were counterstained with Harris’ hematoxylin (HHS16; Sigma-Aldrich, St Louis, MO) and dehydrated using an ethanol gradient and xylene prior to cover-slipping with DPX (06522; Sigma-Aldrich). Images were taken using a Leica (Wetzlar, Germany) DM6000B fluorescence episcope or a Leica SP8 confocal microscope and analyzed with LAS X software. Quantification of DRG soma size Images of H&E stained DRG were processed in Fiji ImageJ for soma size quantification. The freehand selection tool was used to select only the image area containing neural soma. The image was converted to binary, inverted, and auto-thresholded using the minimum method, and soma were separated using watershed. Soma were measured using the particle analyzer with size 150–1000 μm^2 and circularity between 0.4 and 1.0. Quantification of ATXN2 protein knock-down in lumbar spine Sections from L3 and L4 lumbar spine regions were co-stained with antibodies to both ChAT and ATXN2. Mean fluorescence of ATXN2 was quantified exclusively in ChAT-positive cells by a blinded investigator using ImageJ software. To remove background noise, mean fluorescence of extracellular background signal was subtracted from mean fluorescence in ChAT-positive cells. N = 3–6 mice/group, 2–10 images/mouse; graph points depict individual mice normalized to untreated mutant mice. Quantification of motor neurons For upper motor neurons, NeuN-positive cells in the motor cortex were counted using ImageJ, using the Allen mouse brain atlas to identify the M1 primary motor cortex. N = 4–6 mice/group, 1–6 sections/mouse; graph points depict individual brain hemisphere images, which were normalized to the wildtype control from that batch of staining and analyzed using linear mixed models as previously described^[220]8. Images were converted to 8-bit.tif files and triangle threshold was applied to all images. The analyze particles function was used to measure cells larger than 50 microns and with a circularity between 0.2 and 1.0. For lower motor neuron quantification, lumbar regions L3-L6 were counted by two blinded observers and LMNs were identified by their ChAT-positive staining, location in the ventral horn and strict size and morphological criteria (with only diameter >20 µm and polygonal shape counted). Images shown are from the L6 level. N = 4–6 mice/group, 4–10 sections/mouse; graph points depict individual mice normalized to the wildtype control from that batch of staining. Quantification of apoptosis Sections were taken from L3-L6 lumbar spine regions and stained with antibodies to cleaved caspase 3 (CC3). Images shown are from the L3 level where both pathology and rescue were strong. N = 3–7 mice/group at each lumbar level, 2–7 sections/mouse; graph points depict individual mice normalized to the untreated mutant mouse from each batch. CC3-positive cells were hand-counted by a blinded researcher and for each section, the number of positive cells was divided by the area of the section. Quantification of neuroinflammation For brain, sections were selected from the M1 region as outlined above, while for spinal cord, sections were taken from the L6 lumbar region. N = 3–5 mice/group, 3-6 sections/mouse for brain, and N = 5–7 mice/group, 1–4 sections/mouse for spinal cord; graph points depict individual mice normalized to the wildtype control from that batch of staining. Gfap- and Iba1-stained images were analyzed using ImageJ. Images were converted into 8-bit.tif files and a grayness threshold was determined for each batch based on the average wildtype stain presence. The “analyze particles” function was then used to quantify the total area of positive staining by setting the size limit to 0 and using a circularity of 0–1.0. Quantification of phosphorylated TDP-43 Sections were taken from the L6 lumbar region. Color balance threshold was applied to each image, with the threshold value determined by the average of the wildtype group. To determine the total level of phosphorylated TDP-43 in the ventral horn, mean fluorescence was quantified using ImageJ software. Variables selected for measurement included area, integrated intensity, and mean gray value. Multiple areas of the image with no fluorescence were measured as background. The corrected total cell fluorescence (CTCF) was calculated in excel using the following formula: CTCF = Integrated Density − (area of selected cell × mean fluorescence of background readings). N = 4–5 mice/group, 1–5 slides/mouse; graph points depict individual mice normalized to the wildtype control from that batch of staining. Whole transcriptome library preparation and sequencing Total RNA (1ug) was extracted from tissue using Trizol. RNA was quantified by Qubit^TM and RNA integrity (RIN) was assessed by RNA ScreenTape Analysis on an Agilent (Santa Clara, CA) 4200 TapeStation per manufacturer’s protocol. Samples with RIN values < 8 were excluded. cDNA sequencing libraries were enriched for mRNA with the NEBNext® Poly(A) mRNA Magnetic Isolation Module (S7590S; New England Biolabs, Ipswitch, MA) and prepared using NEBNext Ultra II Directional RNA Library Prep Kit with Sample Purification Beads (E7765; New England Biolabs). cDNA libraries were then indexed using the NEBNext Dual Index Kit (E7600S; New England Biolabs). Final cDNA libraries were analyzed by DNA ScreenTape Analysis on an Agilent 4200 TapeStation per the manufacturer’s protocol to determine library size, molar concentration, and purity. Libraries were indexed and pooled at concentrations of 1.5 nM, then run on a NovaSeq 6000 S1 flow cell (Illumina) to a target sequencing depth of 60 million reads per sample using NovaSeq Control Software v1.5. The resulting sequencing reads, in fastq format, were aligned to the Mus musculus genome (GRCm39.104) obtained from ensembl.org. Alignment was performed with the STAR (STAR_2.6.0c) aligner.39. Read counts-per-gene values generated by STAR were used as the basis for differential expression analysis performed using DESeq2 version 1.20.1 (R version 3.6.1). clusterProfiler (version 4.1.4) was used for all functional enrichment analyses. Data visualization was also performed in R and associated modalities within EnhancedVolcano (version 1.9.11), tidyverse (version 1.3.1), dbplyr (version 2.1.1), ggplot2 (version 3.3.5), pheatmap (version 1.0.12), and RColorBrewer (version 1.1.2). Raw and processed next-generation sequencing data have been deposited in the NCBI Gene Expression Omnibus (GEO) database under accession code [221]GSE283631. Generation of Table [222]S1 A literature search was conducted for each of the 23 genes listed in Table [223]S1, using that gene name and any relevant alternative names as search terms and PubMed.gov as the database. Genes relevant to human or other species ALS literature are included in the Supplemental bibliography. iPSC maintenance A male C9orf72-ALS patient iPSC line (CS7VCZiALS-nxx) available through the Answer-ALS Project was obtained via the Cedars-Sinai iPSC repository ([224]https://biomanufacturing.cedars-sinai.org). The cell line is reported to contain >800 repeats in the C9orf72 gene; we confirmed presence of a large repeat expansion using the AmplideX-C9 assay. A dox-inducible transcription factor cassette for lower motor neuron differentiation^[225]25 was inserted by Dr. Esteban Mazzoni at NYU. Karyotyping and CNV analysis were conducted prior to initiation of experiments and cells tested negative for mycoplasma. Cells were maintained in Matrigel (35460; Corning, Corning, NY)-coated plastic dishes in mTeSR Plus Basal Medium (100-0276; StemCell Technologies, Vancouver, BC) at 37 °C and 5% CO[2]. Media was supplemented with ROCK inhibitor Y-27632 (10 uM; 125410; Tocris, Bristol, UK) for the first 24 h after thawing. Neuronal differentiation On day 0, iPSCs were dissociated into single cells with Accutase (A1110501; Gibco, Grand Island, NY) and seeded at 1 × 106 cells/well in a 6-well plate in pre-differentiation media (PDM) (1 x KnockOut DMEM/F-12 [12660012; Gibco], 1 x Glutamax [35050061; Gibco], 1 x MEM NEAA [11140050; Gibco], 1 x N-2 Supplement [17502048; Gibco]). Cell media was supplemented with Y-27632 (10 uM) and doxycycline (2 μg/mL; D3072; Sigma-Aldrich) for 24 h after plating. On day 2, media was changed to PDM with retinoic acid (RA, 0.01uM) and doxycycline (2 μg/mL). On day 3, media was changed to PDM with RA (0.01uM), doxycycline (2 μg/mL), and 5-fluoro-2’-deoxyuridine/Uridine (FdU/Uridine, 10uM each). On day 4, cells were dissociated with Accutase and plated at 5 × 104 cells/well in HCl-treated glass-bottom 24-well plates coated with poly-L-ornithine (PLO) or 2 × 105 cells/well in plastic 6-well plates coated with PLO. Cells were plated in maturation media (MM) (1 x Neurobasal Plus [A3582901; Gibco], 1 x B-27 Plus [A3582801; Gibco], 1 x CultureOne [A3320201; Gibco], 1 x MEM NEAA, 1 x Glutamax) supplemented with doxycycline (2 μg/mL), Y-27632 (10uM), brain-derived neurotrophic factor (BDNF, 10 ng/mL; 450-02; PeproTech, Cranbury, NJ), ciliary neurotrophic factor (CNTF, 10 ng/mL; 450-13; PeproTech), glial cell line-derived neurotrophic factor (GDNF, 10 ng/mL; 450-10; PeproTech), and L-ascorbic acid (200 uM; A8960; Sigma-Aldrich). After 30 min, additional MM was added containing laminin (1 μg/mL). After replating, half media changes were performed on day 7 and day 10 using MM with laminin (1 μg/mL), BDNF (10 ng/mL), CNTF (10 ng/mL), GDNF (10 ng/mL), and ascorbic acid (200 μM). iPSC transduction On day 14, neurons were transduced with PM-AAV9.eGFP or PM-AAV9.miAtxn2 at MOIs of 1 × 104, 5 × 104, 1 × 105, or 5 × 105. Virus was diluted in MM with BDNF (10 ng/mL), CNTF (10 ng/mL), GDNF (10 ng/mL), and ascorbic acid (200 μM) and introduced in a half media change. Media was changed 1x per week following transduction. iPSC immunofluorescence On day 28, iPSC-MNs were washed with DPBS (+/+) (SH30264; Cytiva) and fixed in 4% paraformaldehyde (15711; Electron Microscopy Sciences, Hatfield, PA) and 0.25% glutaraldehyde (16019; Electron Microscopy Sciences) in PHEM buffer (11162; Electron Microscopy Sciences) at pH 7.4 for 10 min. Cells were washed with DPBS (−/−) (SH30028; Cytiva) with Hoechst (1:2500; H3570; Invitrogen) added during the second wash. Cells were permeabilized and blocked in 0.25% Triton-X 100 and 3% goat or donkey serum in DPBS (−/−) for 60 min at RT. Anti-NeuN rabbit monoclonal antibody (1:100; EPR 12763; Abcam) and anti-ChAT goat polyclonal antibody (1:100; AB144P; Millipore) were diluted in blocking buffer and incubated for 60 min at RT. Goat anti-rabbit Alexafluor 568 (A-11011) and Donkey anti-goat AlexaFluor 568 (A-11057) polyclonal IgG antibodies were diluted 1:1000 in blocking buffer and incubated for 1 h at RT. Cells were washed and stored in DPBS (−/−) until imaging. Imaging and GFP quantification Brightfield and GFP images were taken on day 17, 21, 24, and 28 (3, 7, 10, and 14 days post-transduction, respectively). No antibody-based amplification was used, i.e., images are of native GFP fluorescence. Images were taken with a Nikon Eclipse Ti2-E Microscope (Nikon, Melville, NY) at 10x or 40x magnification. Processing of image files was done in FIJI 2 (Version 2.14.0/1.54 f). The Cellpose3 model^[226]55 was used to segment 10x brightfield images for GFP quantification. Cell masks were imported as ROIs into FIJI. Mean gray value was measured for each cell. Un-transduced wells were used to establish a threshold for GFP positivity. Transduction efficiency was calculated as number of GFP positive cells as a percent of total cells. Images from 3 separate wells were quantified for each MOI. Statistics and reproducibility The objective of this study was to determine whether treatment with PM-AAV9.miAtxn2 rescues abnormalities seen in the TAR4/4 mouse model of sporadic ALS, including survival, strength-related behaviors, histopathology, and transcriptomics. Mice studied included male and female TAR4/4 homozygous mice, TAR4 hemizygous mice, and wildtype littermate controls in a series of controlled laboratory experiments. Mice were randomized by litter to receive PM-AAV9.miAtxn2, PM-AAV9.miCtrl, or virus buffer as outlined below and in the manuscript. Treatments were prepared by an independent investigator not involved in the execution of the studies. Injections, all experiments, and all analyses were conducted in a blinded fashion, including the investigators performing the injections or experiments, the investigators analyzing them, and all animal caretakers. Unblinding occurred at the end of the study. Sample sizes for these studies were determined based on predicted effect size and resultant power calculations from a previously published study using the same model and target transcript^[227]8. Treatment endpoints and a detailed analysis plan for each study were determined prior to initiation of the overall study, with the exception that in the case of lumbar neuron quantification, when total lumbar neuron counts were not significantly reduced in the model, additional analysis was done by level. These results are shown in Fig. [228]3. Samples or mice were added to the study if technical errors (e.g., tissue damage during embedding) prevented inclusion of a sample. In some studies, if the magnitude of effect exceeded what was predicted (e.g., in cortical inflammation studies) and statistical significance was achieved early, collection was stopped at that point. All experiments described in this paper used numbers of animals and sections per animal as outlined in each section above and in figure legends. For histology, because the large number of slides required staining in batches, each batch contained representative animals from each treatment group, and results were normalized to wildtype controls contained in that batch. Histology data were excluded only in circumstances where outliers were identified by Tukey’s fences (1.5 IQR, ~0.7%); behavior data were never excluded. Data were plotted and statistical tests were performed using GraphPad Prism. In all cases except for upper motor neuron counts (Fig. [229]S3G), dots on graphs represent individual mice, while for Fig. [230]S3G, graph points depict individual brain hemisphere images which were normalized to the wildtype control from that batch of staining and analyzed using linear mixed models as previously described^[231]8. Except for Fig. [232]S3G, for histology studies the value per mouse was determined by averaging the values of all slides for that mouse. Statistical significance was determined using unpaired two-tailed T-tests when comparing two groups, one-way ANOVA when comparing multiple groups, and two-way ANOVA split by mouse when comparing treatment groups over time or by spinal cord level, using GraphPad Prism. P-values were adjusted for multiple comparisons using the Holm-Sidak method. For neuronal cell counts (Figure [233]S3G), data was analyzed with general linear mixed modeling following the approach outlined by Becker et al.^[234]8 and pairwise multiple comparisons were made using the Holm-Sidak method. A p-value less than 0.05 was considered significant in all studies. Data are represented as mean +/− SEM for all graphs. For bulk RNA-seq analysis, pairwise comparisons were performed using the Wald test (DESeq2 v1.44) and p-values were adjusted for multiple comparisons using the Benjamini-Hochberg method. An adjusted p-value of <0.05 was considered significant, with log2 fold change cut-offs for individual comparisons listed within the results. Principal components analysis identified litter as a covariate, which was included in our model design (~Litter + Genotype_Treatment). For iPSC studies in Fig. [235]8E, F, no statistical method was used as the N was less than 3 per condition. All code related to the statistical analysis of differential gene expression is made available (see Data and materials availability section). Reporting summary Further information on research design is available in the [236]Nature Portfolio Reporting Summary linked to this article. Supplementary information [237]Supplementary Information^ (5MB, pdf) [238]Reporting Summary^ (94.7KB, pdf) [239]Transparent Peer Review file^ (267.6KB, pdf) Source data [240]Source Data^ (37.5MB, xlsx) Acknowledgements