Author Response: RNA-binding Deficient TDP-43 Drives Cognitive Decline in a Mouse Model of TDP-43 Proteinopathy
Julie Necarsulmer,Jeremy M. Simon,Baggio Evangelista,Youjun Chen,Xu Tian,Sara Nafees,Ariana B Marquez,Huijun Jiang,Ping Wang,Deepa Ajit,Viktoriya D. Nikolova,Kathryn M. Harper,Jennifer Ezzell,Feng‐Chang Lin,Adriana S. Beltrán,Sheryl S. Moy,Todd J. Cohen
DOI: https://doi.org/10.7554/elife.85921.3.sa4
2023-01-01
Abstract:Full text Figures and data Side by side Abstract eLife assessment Introduction Results Discussion Methods Data availability References Peer review Author response Article and author information Metrics Abstract TDP-43 proteinopathies including frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS) are neurodegenerative disorders characterized by aggregation and mislocalization of the nucleic acid-binding protein TDP-43 and subsequent neuronal dysfunction. Here, we developed endogenous models of sporadic TDP-43 proteinopathy based on the principle that disease-associated TDP-43 acetylation at lysine 145 (K145) alters TDP-43 conformation, impairs RNA-binding capacity, and induces downstream mis-regulation of target genes. Expression of acetylation-mimic TDP-43K145Q resulted in stress-induced nuclear TDP-43 foci and loss of TDP-43 function in primary mouse and human-induced pluripotent stem cell (hiPSC)-derived cortical neurons. Mice harboring the TDP-43K145Q mutation recapitulated key hallmarks of FTLD, including progressive TDP-43 phosphorylation and insolubility, TDP-43 mis-localization, transcriptomic and splicing alterations, and cognitive dysfunction. Our study supports a model in which TDP-43 acetylation drives neuronal dysfunction and cognitive decline through aberrant splicing and transcription of critical genes that regulate synaptic plasticity and stress response signaling. The neurodegenerative cascade initiated by TDP-43 acetylation recapitulates many aspects of human FTLD and provides a new paradigm to further interrogate TDP-43 proteinopathies. eLife assessment Necarsulmer et al describe an interesting new mouse model of TDP-43 proteinopathy in which gene editing was used to introduce a K145Q acetylation-mimic mutation previously shown to impair RNA-binding capacity and induce downstream misregulation of target genes. Mice homozygous for this mutation are convincingly shown to display cognitive/behavioral impairment, TDP-43 phosphorylation and insolubility, and changes in gene expression and splicing. This novel mouse model replicates some important hallmarks of human frontotemporal lobar degeneration and will be an important contribution to the field. https://doi.org/10.7554/eLife.85921.3.sa0 About eLife assessments Introduction TDP-43 proteinopathies are characterized by the dysfunction and aggregation of transactivation response element DNA-binding protein of 43 kDa (TDP-43), with ~95% of all amyotrophic lateral sclerosis (ALS) and ~50–60% of all frontotemporal lobar dementia (FTLD-TDP) cases harboring TDP-43 pathology (Neumann et al., 2006; Neumann et al., 2007; Cairns et al., 2007; Hogan et al., 2016). There is significant neuropathologic and clinical overlap between FTLD and ALS with many individuals developing a mixed phenotype, providing strong evidence for a common FTLD/ALS spectrum of disorders (Burrell et al., 2016; Geser et al., 2010; Burrell et al., 2011; Giordana et al., 2011; Geser et al., 2009). It is also notable that TDP-43 pathology is abundant in other sporadic neurodegenerative diseases including Alzheimer disease (AD) (Meneses et al., 2021; Tomé et al., 2020), Limbic-Predominant Age-related TDP-43 Encephalopathy (LATE) (Besser et al., 2020; Nelson et al., 2019), and Parkinson’s disease (PD) (Poulopoulos et al., 2012; Nakashima-Yasuda et al., 2007). The clinical and neuropathological overlap suggests that common pathogenic mechanisms may link TDP-43 to neurodegeneration (Gao et al., 2018; de Boer et al., 2021). However, modeling sporadic TDP-43 pathogenesis has been challenging since its expression levels are tightly regulated (Budini and Buratti, 2011; Ayala et al., 2011), which has precluded a clear separation of TDP-43 disease-related dysfunction from general toxicity resulting from TDP-43 over- or under-expression (Xu et al., 2010; Igaz et al., 2011; Yang et al., 2014; Kraemer et al., 2010). Current knock-in models using TDP-43 disease-causing mutations (Fratta et al., 2018; White et al., 2018; Huang et al., 2020; Stribl et al., 2014; Ebstein et al., 2019) provide valuable insights but may be limited in their application to sporadic disease (Fratta et al., 2018; White et al., 2018; Huang et al., 2020; Stribl et al., 2014; Ebstein et al., 2019). Under normal physiological conditions, TDP-43 resides in the nucleus to control RNA processing (RNA splicing, transport, and stability) and gene transcription (Buratti and Baralle, 2010; Tollervey et al., 2011; Cohen et al., 2011). Structurally, nuclear retention is primarily mediated by an N-terminal nuclear localization sequence (NLS) through interactions with α1/β-importins (Doll et al., 2022; Pinarbasi et al., 2018), and association with nucleic acids is mediated by two tandem RNA recognition motifs (RRM1/RRM2) (Kuo et al., 2014; Lukavsky et al., 2013), however there is interplay between nucleic acid binding and nuclear localization (Duan et al., 2022; Ayala et al., 2008). The C-terminal glycine-rich domain (also termed the intrinsically disordered or low complexity domain) mediates protein–protein interactions (Ayala et al., 2008; Buratti and Baralle, 2012; Budini et al., 2012) and harbors most, but not all, familial TARDBP mutations that are causative for FTLD/ALS (Pesiridis et al., 2009; Sreedharan et al., 2008). In sporadic and most familial TDP-43 proteinopathies, TDP-43 undergoes nuclear depletion and concomitant nuclear or cytoplasmic accumulation and aggregation (Kawakami et al., 2019; Neumann, 2009; Mackenzie and Neumann, 2016). Both TDP-43 loss-of-function (e.g., aberrant cryptic splicing) and gain-of-function (e.g., aggregate-induced toxicity) mechanisms have been proposed as drivers of TDP-43 pathogenesis (Cascella et al., 2016; Diaper et al., 2013; Lee et al., 2011; Vanden Broeck et al., 2014). How TDP-43 becomes dysfunctional in sporadic disease remains unresolved, however, aberrant TDP-43 post-translational modifications (PTMs), such as phosphorylation, acetylation, and ubiquitination may play a role. PTMs modulate TDP-43’s biochemical properties leading to conformational changes, modulation of nucleic acid-binding affinity, regulation of liquid–liquid phase separation, and propensity to form insoluble TDP-43 aggregates, all of which are disease-associated phenomena (François-Moutal et al., 2019; Sternburg et al., 2022; Buratti, 2018). Among the various PTMs, TDP-43 acetylation at lysine residue 145 (K145) within RRM1 has emerged as a critical regulator of both loss- and gain-of-function toxicity (Cohen et al., 2015; Wang et al., 2017). Acetylated TDP-43 is found within TDP-43 inclusions of sporadic ALS spinal cord motor neurons but not age-matched control tissue (Cohen et al., 2015). Inclusions in FTLD cortex are largely composed of C-terminal fragmented TDP-43 lacking the K145 residue (Igaz et al., 2008; Chhangani et al., 2021), precluding an assessment of Ac-K145 in FTLD patients. However, TARDBP mutations that disrupt RNA binding, and thereby may act in a similar manner to TDP-43 acetylation, have been identified in FTLD-TDP patients (e.g., P112H and K181E) (Agrawal et al., 2021; Chen et al., 2019), supporting a pathogenic role for altered nucleic acid binding in disease. Mimicking TDP-43 acetylation with a lysine-to-glutamine substitution (TDP-43K145Q) is sufficient to neutralize the positive charge, disrupt RNA binding, and induce several hallmarks of TDP-43 pathology in vitro (Cohen et al., 2015; Wang et al., 2017), supporting a model whereby TDP-43 acetylation drives both loss-of-function (e.g., RNA-binding deficiency) and gain-of-function (e.g., aggregation) toxicity. Here, we used CRISPR/Cas9 genome editing to introduce a K145Q substitution into the endogenous mouse Tardbp gene, thereby generating acetylation-mimic TDP-43K145Q knock-in mice, which enabled us to investigate the pathophysiological impacts of an aberrant TDP-43 PTM while leaving native upstream and downstream genomic elements intact. Using mouse cortical neurons, human hiPSC-derived cortical neurons, and aged cohorts of TDP-43K145Q homozygous mice, we found that acetylation-mimic TDP-43K145Q-induced nuclear TDP-43 foci and cytoplasmic TDP-43 accumulation, which coincided with several disease-associated and loss-of-function measures including widespread transcriptome and splicing alterations. Finally, we observed prominent FTLD-like cognitive and behavioral deficits in acetylation-mimic TDP-43 mice that correlated with biochemical and splicing alterations in affected brain regions. Our study supports lysine acetylation of TDP-43 as a driver of dysfunction in sporadic TDP-43 proteinopathies. Results Mouse neurons expressing TDP-43K145Q undergo stress-dependent formation of nuclear TDP-43 foci and loss of TDP-43 splicing function We originally showed that TDP-43 acetylation can promote RNA-binding deficiency, aggregation, and pathology (Cohen et al., 2015; Wang et al., 2017). We sought to expand these findings by exploring the behavior of acetylation-mimic TDP-43 variants in primary murine cortical neurons. We employed lentiviral vectors that encode either wild-type (TDP-43wt), acetylation-deficient (TDP-43K145R), and acetylation-mimic (TDP-43K145Q) variants to overexpress TDP-43 species in neurons, and then examined their subcellular localization by immunofluorescence microscopy. In the absence of acute cellular stress, most neurons overexpressing TDP-43K145Q showed distinct nuclear foci that were more prominent compared to TDP-43wt or TDP-43K145R constructs (Figure 1A, B). When neurons were exposed to an acute oxidative stressor (200 µM sodium arsenite), a sensitizing agent commonly used to enhance TDP-43 dysfunction (Dewey et al., 2011; Colombrita et al., 2009; Gasset-Rosa et al., 2019; Cohen et al., 2012), there was a significant increase in TDP-43 foci formation with all variants (Figure 1A, C). The response in neurons expressing TDP-43K145Q was very robust, resulting in the formation of numerous large, bright TDP-43-positive foci, as well as smaller TDP-43-positive nuclear foci. These aberrant TDP-43 structures were absent from cells expressing TDP-43wt or acetylation-null TDP-43K145R, indicating that acetylation-mimic TDP-43K145Q alters TDP-43 conformation within the nucleus and sensitizes neurons to oxidative stress-induced foci formation. By coupling high content wide-field microscopy with quantitative image analysis, we observed a threefold increase in TDP-43 foci formation in neurons expressing TDP-43K145Q (Figure 1C). Figure 1 Download asset Open asset Ectopic expression of acetylation-mimic TDP-43K145Q, an RNA-binding deficient mutant, shows accelerated formation of stress-induced nuclear TDP-43 foci. (A) Representative immunofluorescence images of TDP-43 in DIV14 mouse primary cortical neurons overexpressing TDP-43wt, TDP-43K145Q, or TDP-43K145R after vehicle or 200 µM NaAsO2 treatment followed by labeling of TDP-43 (green), NeuN (magenta), and DAPI (blue). Arrows highlight nuclei with TDP-43+ foci. Scale bar = 20 µm. (B) Quantification of percentage of neurons with TDP-43+ foci in vehicle-treated neurons. (C) Quantification of the average number of TDP-43+ foci per neuron. Data shown as Superplots (Lord et al., 2020); solid color bordered symbols and error bars indicate mean value of each biological replicate ± standard error of the mean (SEM); semi-transparent datapoints represent the average value per neuron in a single field of view, 10–110 neurons per field, 48 fields across n = 2–4 biological replicates. Statistical analysis completed using a linear mixed-effect model. Statistical significance is represented by asterisks *p < 0.05, **p < 0.01, ***p < 0.001. DIV = day in vitro. TDP-43 overexpression can result in general toxicity and altered TDP-43 function, depending on the duration and the extent of overexpression (Xu et al., 2010; Ash et al., 2010; Yang et al., 2022). To avoid confounding non-specific toxicity, we transitioned to a more physiologically relevant model to further elucidate the impact of RNA-binding deficient acetylation-mimic TDP-43. We employed CRISPR-Cas9 mutagenesis to introduce a single amino acid substitution at position 145 (K145Q) into the endogenous mouse Tardbp locus, thereby generating TDP-43K145Q knock-in mice (Figure 2—figure supplement 1B). By targeting the native mouse gene, we avoided both TDP-43 overexpression and disruption of the Tardbp untranslated regions (Budini and Buratti, 2011 ). A TDP-43K145Q founder line was propagated as heterozygotes and continually re-sequenced to confirm retention and propagation of the K145Q substitution (Figure 2—figure supplement 1C, D). Both heterozygous and homozygous TDP-43K145Q mice were born at normal mendelian frequencies and showed no obvious developmental defects. We first investigated the effects of TDP-43K145Q expression in neurons in vitro by isolating and culturing primary cortical neurons from homozygous TDP-43K145Q mice, hereafter referred to as TDP-43KQ/KQ mice and compared them to TDP-43wt-derived neurons. Exposing neurons to acute oxidative stress induced more abundant TDP-43-positive nuclear foci in acetylation-mimic TDP-43KQ/KQ neurons than in TDP-43wt neurons (Figure 2A, B). Quantitative image analysis also revealed variable levels of nuclear clearing and cytoplasmic mislocalization of TDP-43 in TDP-43KQ/KQ neurons, however, no statistically significant differences in TDP-43 localization were found between acetylation-mimic and TDP-43wt neurons (Figure 2A, C). Because TDP-43 foci formation is associated with loss-of-function defects (Garcia Morato et al., 2022; Mann and Donnelly, 2021; Yu et al., 2021), we next investigated if TDP-43 function was impaired by evaluating endogenous targets of TDP-43 activity. We observed a trend toward increased Tardbp mRNA in TDP-43KQ/KQ neurons at DIV14 (Figure 2D), which was suggestive of TDP-43 loss of function and auto-regulation. We therefore investigated sortillin-1 (Sort1) mRNA splice variants, as a more sensitive endogenous indicator of loss of TDP-43-dependent splicing function. Functional nuclear TDP-43 results in the production of a mature spliced Sort1 mRNA transcript, however, in the setting of TDP-43 depletion or loss of function, TDP-43 is unable to repress the inclusion of exon 17b, generating a longer Sort1+ex17b transcript variant (Prudencio et al., 2012; Tann et al., 2019). RT-qPCR analysis revealed a significant deficit in normal TDP-43 splicing function, with nearly a 10-fold increase in the ratio of Sort1+ex17b to Sort1-WT mRNA, without significant changes to total Sort1 mRNA levels in TDP-43KQ/KQ neurons compared to controls (Figure 2E, F). Overall, these results indicate that a single endogenously expressed acetylation-mimic TDP-43K145Q mutation can sensitize TDP-43 to conformational changes that impair its normal splicing function in mouse neurons. Figure 2 with 1 supplement see all Download asset Open asset An endogenously encoded acetylation-mimic TDP-43K145Q mutation causes altered TDP-43 localization and functional splicing deficits in mouse primary cortical neurons. (A) Representative images of primary cortical neurons derived from TDP-43wt or TDP-43KQ/KQ mice that were treated with vehicle or 200 µM NaAsO2 at DIV14 and immunolabeled for endogenous TDP-43 (green), NeuN (magenta), and DAPI (blue). Quantification of the number of TDP-43+ foci (B) and the cytoplasmic:nuclear ratio of TDP-43 fluorescence intensity (C) in TDP-43KQ/KQ compared to TDP-43wt neurons. RT-qPCR analysis of DIV14 and DIV28 untreated mouse primary cortical neurons using primers specific for mouse (D) Tardbp [F(1,8) = 3.034, p = 0.1197]; and Sort1 splice variants: (E) ratio Sort1+ex17b:Sort1-WT [F(1,8) = 23.02, p = 0.0014] and (F) total Sort1 mRNA [F(1,8) = 5.086, p = 0.0541]. (B, C) Data shown as Superplots (Lord et al., 2020); solid color bordered symbols and error bars indicate mean value of each biological replicate ± standard error of the mean (SEM); semi-transparent datapoints represent the average value per neuron in a single field of view, 10–110 neurons per field, 72 fields across n = 2–4 biological replicates; one color represents one biological replicate. Statistical analysis performed using a linear mixed-effect model. (D–F) Analysis by two-way analysis of variance (ANOVA) followed by Šídák’s multiple comparisons testing. F statistics represent main effect of genotype. Statistical significance is represented by asterisks, *p < 0.05, **p < 0.01, ****p < 0.0001, ns = not significant. Acetylation-mimic TDP-43K145Q alters TDP-43 function in human neurons To assess this model’s relevance to human neurons, we used CRISPR/Cas9 to generate a panel of hiPSC lines harboring homozygous acetylation-mimic TDP-43 (TDP-43K145Q.12 and TDP-43K145Q.18), acetylation-deficient TDP-43 (TDP-43K145R.2 and TDP-43K145R.12), or unmodified TDP-43 (isogenic control, TDP-43wt) (Figure 3—figure supplement 1). We confirmed appropriate editing of the TARDBP gene in each line via Sanger sequencing (Figure 3—figure supplement 1B), verified the pluripotency of the hiPSC clones (Figure 3—figure supplement 1C–E), and then differentiated each of the lines into mature cortical neurons (Figure 3—figure supplement 2). We then used immunofluorescent labeling and confocal microscopy to qualitatively assess the distribution and morphology of TDP-43 protein within neurons, both with and without an acute exposure to sodium arsenite. Untreated hiPSC-derived TDP-43K145Q cortical neurons were morphologically identical to TDP-43wt and TDP-43K145R neurons and showed similar patterns of TDP-43 localization (Figure 3A). All hiPSC-derived lines showed a granular nuclear TDP-43 localization pattern under normal conditions, consistent with physiologic de-mixing of nuclear TDP-43 (Gasset-Rosa et al., 2019). Following acute oxidative stress, however, TDP-43K145Q neurons showed apparent TDP-43 nuclear clearing and the formation of large, intensely labeled TDP-43-positive foci (Figure 3A). In comparison, cortical neurons expressing TDP-43wt or TDP-43K145R maintained nuclear TDP-43 and formed small stippled TDP-43 puncta. We note that TDP-43 nuclear clearing and foci formation was more robust in hiPSC-derived neurons compared to mouse neurons, suggesting human neurons may be more sensitive to TDP-43 RNA-binding deficiency. Similar to mouse neurons, hiPSC-derived TDP-43K145R neurons were indistinguishable from TDP-43wt neurons, supporting acetylation-induced charge neutralization as a driver of TDP-43 loss of function rather than an inherent effect of mutation at the K145 locus. Figure 3 with 2 supplements see all Download asset Open asset RNA-binding deficient TDP-43K145Q in human-induced pluripotent stem cell (iPSC)-derived mature cortical neurons alters TDP-43 distribution and causes disease-relevant splicing dysregulation. (A) Representative confocal images of CRISPR-modified human iPSC-derived cortical neurons harboring homozygous TDP-43K145Q (clonal lines 12, 18) or TDP-43K145R (clonal lines 2, 12) knock-in mutations or an isogenic wild-type TDP-43wt control lacking TARDBP modifications. Differentiated cortical neurons were treated with vehicle or 200 µM NaAsO2 and then immunolabeled for endogenous TDP-43 (green), Map2 (magenta), and DAPI (blue). Scale bars = 20 µm. Arrows highlight nuclei with few, bright TDP-43+ foci. Arrowheads highlight nuclei with relative depletion of TDP-43 intensity. RT-qPCR analysis of hiPSC-derived mature cortical neuron samples using primers specific for (B) human Tardbp [F = 43.79, p < 0.0001], (C) SORT1+ex17b, (D) SORT1-WT [F = 2.975, p = 0.0516], (E) total SORT1 mRNA [F = 3.461, p = 0.0321], (F) truncated STMN2 [F = 79.06, p < 0.0001], (G) STMN2 WT [F = 63.93, p < 0.0001], (H) UNC13A cryptic exon, and (I) UNC13A WT mRNA [F = 2.116, p = 0.1262]. RT-qPCR results compared via ordinary one-way analysis of variance (ANOVA) (F statistics and p-values shown in brackets above) followed by Tukey’s multiple comparisons testing. Statistical significance of multiple comparisons testing is represented by asterisks *p < 0.05, ****p < 0.0001. †Graphs are provided for visualization purposes only, because non-detectable levels of UNC13A cryptic exon- and SORT1-ex17b-containing transcripts in some or all control TDP-43wt and TDP-43K145R neurons prevented statistical comparisons among groups and therefore no statistical significance was reported. We next asked whether hiPSC-derived TDP-43K145Q cortical neurons show more robust functional deficits compared to the mouse neuron data above. We again employed RT-qPCR to measure levels transcripts associated with TDP-43 loss of function, beginning with TARDBP as an indicator of impaired TDP-43 autoregulation. In contrast to mouse neurons, we observed an approximately twofold increase TARDBP mRNA in TDP-43K145Q.12 and TDP-43K145Q.18 neurons and a slight but significant reduction in TARDBP mRNA in TDP-43K145R.12 neurons (Figure 3B). We then assessed TDP-43-dependent splicing activity using primers specific for human SORT1 splice variants, whose regulation by TDP-43 is conserved in mouse and humans (Prudencio et al., 2012; Tann et al., 2019). The aberrant SORT1+ex17b splice variant was undetectable in TDP-43wt neurons, but was present in 60% of TDP-43K145Q.12 and 100% of TDP-43K145Q.18, TDP-43K145R.2, and TDP-43K145R.12 neurons (Figure 3C). In samples confirmed to have detectable SORT1+ex17b transcript, levels were, on average, considerably higher levels in TDP-43K145Q compared to TDP-43K145R neurons. Appropriately spliced SORT1-WT and total SORT1 transcript levels were comparable across all five lines, with only a slight increase in total SORT1 transcript in TDP-43K145Q.12 neurons (Figure 3D, E). We conclude that SORT1+ex1b is a low-abundance transcript that accumulates in TDP-43K145Q neurons. We also examined neurons for the presence of truncated STMN2 and UNC13A cryptic exon mRNA, which are implicated as ALS–FTLD biomarkers and/or risk genes (Brown et al., 2022; Prudencio et al., 2020; Melamed et al., 2019; Ma et al., 2022). Strikingly, truncated STMN2 mRNA was approximately 100-fold higher in both TDP-43K145Q.12 and TDP-43K145Q.18 neuron lines, respectively, compared to TDP-43wt neurons, while no difference from WT was found in TDP-43K145R neurons. (Figure 3F). The unperturbed STMN2 splice variant (STMN2 WT) was reduced about 50% in both acetylation-mimic TDP-43K145Q lines but unchanged in TDP-43K145R.2 neurons (Figure 3G). We also noted a subtle increase in STMN2 WT mRNA in TDP-43K145R neurons, suggesting the fully deacetylated state, achieved by acetylation-deficient mimics, may augment STMN2 levels. RT-qPCR analysis of cryptic exon-included UNC13A splice variant (UNC13A cryptic exon) failed to detect the abnormal splice product in TDP-43wt and TDP-43K145R.2 lines, with only low levels observed in some TDP-43K145R.12 samples. In contrast, both TDP-43K145Q lines dramatically accumulated the UNC13A cryptic exon containing mRNA (Figure 3H). The mRNA levels of the typical splice variant (UNC13A WT) were similar across all five lines (Figure 3I), indicating that the lack of detectable UNC13A cryptic exon mRNA in WT and acetylation-deficient neuron lines was not a result of reduced UNC13A expression, but rather is likely specific to a lack of cryptic splicing events. Taken together, these results demonstrate that endogenously expressed acetylation-mimic TDP-43K145Q alters TDP-43 localization and conformation in response to stress and impairs splicing in a disease-relevant manner in human iPSC-derived neurons. TDP-43 acetylation-mimic mice develop age-dependent cognitive and behavioral defects To evaluate TDP-43 acetylation-mimic mice, we aged the animals and performed an extensive battery of behavioral analysis to assess cognitive and motor function, which reflect impairments that are commonly found in patients with TDP-43 proteinopathies (Geser et al., 2010). To be sufficiently powered to detect small differences in behavioral phenotypes, we initially focused on TDP-43wt and homozygous TDP-43KQ/KQ mice. Moreover, since there were no significant differences between males and females in any behavioral analyses described below, we pooled both sexes into either WT or TDP-43KQ/KQ groups. At 12 months old, TDP-43KQ/KQ mice showed significant reduction of body weight compared to WT littermates, and this difference was maintained until end point analysis at 18 months old (Figure 4A). Evaluation of exploratory activity and locomotion in an open-field test demonstrated that TDP-43KQ/KQ mice spend significantly more time in the center region (Figure 4B), with no differences in total distance traveled (Figure 4—figure supplement 3A), indicative of decreased anxiety-like behavior (Carola et al., 2002; Seibenhener and Wooten, 2015). Acoustic startle testing revealed impaired prepulse inhibition (PPI) in TDP-43KQ/KQ mice at 12 months old (Figure 4—figure supplement 2A), indicative of deficits in sensorimotor gating, a form of inhibitory behavioral control (Gómez-Nieto et al., 2020; Mena et al., 2016), which is a phenomenon that can be observed in early dementia (Ueki et al., 2006). In 18-month-old animals, evaluation of acoustic startle response and PPI was confounded by hearing impairment (Figure 4—figure supplement 2B), however the altered activity reflected by increased time in the center region of an open-field test was maintained at this age (Figure 4B). Thus, consistent patterns of behavioral disinhibition and reduced anxiety-like behavior were apparent in TDP-43KQ/KQ mice over time. Figure 4 with 3 supplements see all Download asset Open asset TDP-43KQ/KQ mice develop age-dependent cognitive and behavioral defects. (A) Body weight of TDP-43wt and TDP-43KQ/KQ mice in males [left panel; F(1,48) = 17.52, p = 0.0001] and females [right panel; F(1,36) = 14.17, p = 0 .0006] at different ages. (B) Quantification of time in the center region of an open field in mice at 12 months [left panel; F(1,20) = 6.118, p = 0.0225] and 18 months [right panel; F(1,17) = 9.622, p = 0.0065]. (C) Quantification of time spent frozen (immobile) following context-dependent conditioned fear testing at 18 months old [right panel; F(1,17) = 5.402, p = 0.0328]. (D) Quantification of time spent frozen following cue-dependent conditioned fear testing at 12 months old [left panel; F(1,20) = 6.136, p = 0.0223] and 18 months old. Filled bars represent presence of auditory cue (tone) and period of statistical analysis. Morris Water Maze (MWM) analysis displaying time to find a hidden platform (escape latency), quantified as daily trial means per animal (E) or average across all days (F). Quantification of escape latencies during MWM reversal learning trials in daily trials (G) [F(1,16) = 5.273, p = 0.0355] and comparing averages per mouse across all days (H). Bar and scatter plots shown as mean ± standard deviation (SD). Box and whiskers show line at median, ‘+’ at mean, and whiskers run min to max. (A) Two-way analysis of variance (ANOVA) followed by Šídák’s multiple comparisons test. (B–D, E, G) Two-way repeated measures ANOVA followed by Holm–Šídák’s multiple comparisons tests; F statistics and p-values in legend represent main effect of genotype. (F, H) Unpaired Student’s t-test. Sample sizes as follows unless otherwise indicated: 12-month TDP-43wt n = 15; 12-month TDP-43KQ/KQ n = 7; 1-month TDP-43wt n = 10; 18-month TDP-43KQ/KQ n = 9; one 18-month TDP-43KQ/KQ extreme outlier removed for MWM analysis (E, F). Statistical significance is represented by asterisks *p < 0.05, **p < 0.01, ***p < 0.001; ns = not significant. Further statistical information is located in Figure 4—source data 1 file. Figure 4—source data 1 Complete statistical results and information for quantitative elements shown in Figure 4. https://cdn.elifesciences.org/articles/85921/elife-85921-fig4-data1-v1.xlsx Download elife-85921-fig4-data1-v1.xlsx We next performed contextual and cue-dependent fear conditioning as an index of hippocampal and cortical function (Chen et al., 1996; Curzon et al., 2009; Phillips and LeDoux, 1992; Kim and Jung, 2006; Marschner et al., 2008). Context-dependent fear testing revealed reduced freezing times in TDP-43KQ/KQ mice, with trends observed at 12 months old and significant deficits appearing at 18 months (Figure 4C). Similarly, auditory cue-dependent fear testing revealed significant impairments in associative cue learning in TDP-43KQ/KQ mice at 12 months of age (Figure 4D), a behavior thought to be mediated by the amygdala and higher-order cortical regions important in inhibitory control (Sierra-Mercado et al., 2011). As mentioned above, general auditory defects in both genotypes at 18 months of age confounded interpretations of any cue-dependent learning deficits at this advanced age (Figure 4D). Morris Water Maze testing was used to evaluate swimming ability and spatial learning (Vorhees and Williams, 2006), which showed equivalent swim speeds, suggesting no motor impairments in TDP-43KQ/KQ mice at 18 months of age (Figure 4—figure supplement 2D). While assessment of spatial learning showed a trend toward impaired acquisition learning (Figure 4E, F), we observed more prominent defects in reversal learning after moving the location of the platform, as determined by significant delays in escape latency. Our findings support deficits in cognitive flexibility in TDP-43KQ/KQ mice (Figure 4G, H; Nicholls et al., 2008). We also analyzed cognitive impairments in heterozygous TDP-43wt/KQ mice, which showed a mild intermediate phenotype (Figure 4—figure supplement 1). To determine any ALS-like motor phenotypes, we assessed motor function and were surprised to find no overt signs of motor impairment in TDP-43KQ/KQ mice at 18 months of age.