Optimization of the high-throughput screen on human iPSC-derived neurons
We used human neurogenin 2 (NGN2)-driven iPSC-derived neurons (NGN2-iNs) as a physiologically relevant cell-based screening platform to investigate neuronal miRNome and focused on an essential neuron-enriched and neuroprotective miR-132. iPSC lines generated from donors were utilized for direct differentiation through NGN2 overexpression into excitatory neurons based on established protocols (Fig. 1A) (21). These cells closely mimic the transcriptome and function of human neurons ex vivo and can be scaled and reproducibly employed in multiple assays (21). Among 36 NGN2-iN lines obtained from the Religious Orders Study/Memory and Aging Project (ROS-MAP) cohort, 25 lines from donors without cognitive impairment were considered (Fig. S1A). The transcriptomes of these lines were previously profiled (21). The BR43 line was selected for the screen based on its median expression of major miR-132 targets, including GSK3β, EP300, RBFOX1, CAPN2, FOXO3, TMEM106B, and MAPT (Fig. S1B). BR43 NGN2-iNs also had the lowest variation of baseline miR-132 expression among the replicate cultures and exhibited miR-132 upregulation by the known inducers BDNF and forskolin (Fig. S1C).
Several steps of NGN2-iN culture and RNA collection were optimized for HTS to maximize neuronal health, lysing efficiency, and RNA yield. The protocol was tested for compatibility with small RNA-seq using the RealSeq ultra-low input system, long RNA RT-qPCR using the PrimeScript system, and small RNA RT-qPCR using the miRCURY system (S1D, E), supporting its application in diverse quantitative RNA-based assays.
Screen for small molecule regulators of microRNAs
Day 4 NGN2-iNs were plated onto 25 Matrigel-coated 96-well plates and differentiated into neurons, as verified by NeuN and Tau expression (Fig. 1A). On day 19, the Selleckchem library (N = 1,902 compounds), a diverse library of bioactive molecules, was pin-transferred into plates to achieve 10 µM final concentration. DMSO (0.1% final concentration) and forskolin (10 µM) were used as the negative and positive controls, respectively. NGN2-iNs were imaged to monitor neuronal health 24h later, followed by direct lysis to release RNA. Among all wells with test compounds, 324 (17.0%) were excluded because of cell death, neurite degeneration, loss of cells during washes, or enrichment of astrocytes. RNA lysates of the remaining wells were used for RealSeq small RNA library preparation designed for ultra-low input without RNA purification (22). RealSeq libraries from each set of four 96-well culture plates were indexed with 384 multiplex barcodes and pooled for deep sequencing. After miRNA annotation, wells with less than 1,000 total annotated read counts were excluded from further analysis (N = 169, 10.7%). On average, 55,529 miRNA reads were counted per sample, and 455, 240, 182, and 64 miRNA species per sample were detected with minimal read counts of 1, 5, 10, and 100, respectively (Fig. 1B). Numerous neuronal miRNAs, such as miR-26a, miR-7, miR-191, miR-124, and miR-9/9* were abundant in DMSO-treated control NGN2-iNs (Fig. 1C). miR-132 was consistently detected and ranked among the 30 most abundant miRNAs. We further determined the top housekeeping neuronal miRNAs by calculating the coefficient of variation (COV) for each miRNA within each batch of RNA-seq and identified the miRNAs with the smallest COVs, including miR-103a/b, miR-107, and miR-191 (Fig. 1D). Figure 1E showed the miR-132 waterfall plot for 221 compounds in a 384-well plate.
miRNome-scale HTS dataset as a resource to study miRNA-small molecule relationships
Across 6 batches, we obtained miRNA profiles for 1441 samples, including 46 DMSO samples, 26 forskolin samples, and 1369 small molecule compounds (Table S1-2). Each compound was annotated with a summary, including clinical indication if in clinical use (N = 745), pathway, blood-brain barrier permeability, and target or compound class. As library preparation and sequencing for different 384-well plates were carried out on different days, a significant batch effect was observed (Fig. S2). To reduce batch-specific effects and allow for comparison across batches, the expression level of each miRNA was normalized to the DMSO controls in the same batch for the analyses done in Fig. 2 (Table S3).
The dataset can be utilized to study miRNAs and small molecule compounds from various angles. First, it can be used to explore small molecule compounds that modulate a particular miRNA of interest. For example, as miR-132 is downregulated in neurodegenerative diseases, upregulating miR-132 may provide therapeutic effects. Notably, compounds approved for treating neurological diseases appear to mildly upregulate miR-132, suggesting that miR-132 may partly mediate their efficacy (Fig. 2A). More specifically, miR-132 is upregulated by compounds classified as Na, K-ATPase inhibitors, forskolin (positive control), and antiviral (Fig. 2B). Conversely, miR-26b was shown to promote apoptosis in neurons, and reducing miR-26b provided neuroprotection (23). In contrast to miR-132, miR-26b is mildly downregulated by compounds used for treating neurological diseases and inflammation (Fig. 2C). Compounds classified as anesthetics, selective serotonin uptake inhibitors (SSRI), and SIRT inhibitors appear likely to downregulate miR-26b (Fig. 2D). Other neuronal miRNAs of interest and candidate compounds that may regulate them are listed in Table S4.
Second, the dataset can be used to explore the effects of a class of compounds on the miRNome. For example, COX-1 and − 2 inhibitors are commonly used to treat pain and inflammation. Compared to DMSO controls, COX inhibitors generally downregulated miR-26b, which was shown to promote inflammation (24), and upregulated various miRNAs, including miR-125a/b (25), miR-197 (26), and miR-181(27) which were shown to be anti-inflammatory (Fig. 2E). Another example is vascular endothelial growth factor receptor (VEGFR) inhibitors which are used to treat various cancers, but their impact on the nervous system is not well-understood. VEGFR inhibitors commonly downregulated miR-7, the third most abundant miRNA in the screen, and miR-376a, b, and c (Fig. 2F). Whether these changes contribute to the anticancer effects or possible adverse neurological effects of VEGFR inhibitors can be further studied.
Last but not least, the dataset can be utilized to study the relationship between miRNAs. Figure 2G shows the correlation map among the 20 most abundant miRNAs. For example, three transcripts, miR-103a, miR-103b, and miR-107, show a high correlation, probably because they belong to the same family. Figure 2H shows miRNAs with high positive or negative correlations with miR-132 expression. miR-132 shows the strongest negative correlation with miR-26b and the let-7 family, which were previously reported to be neurotoxic (28), suggesting that distinct populations of miRNAs are associated with neuroprotection and neurotoxicity, respectively.
Screen validation: cardiac glycosides upregulate miR-132 transcriptionally and specifically
We selected miR-132, a well-known neuroprotector, as the paradigm to show clinical utilization of the miRNome-scale HTS dataset. To filter compounds that may upregulate its expression, we used miR-132 plate rank as the primary criterion and adjusted with secondary criteria, including clinical approval, BBB penetrance, clinical trials, published data on neuroprotective effects, and effects on other miRNAs. We treated DIV14 primary rat cortical neurons and DIV21 human NGN2-iNs with 10 µM of 44 selected compounds and monitored miR-132 expression by RT-qPCR. 12 and 10 compounds significantly upregulated miR-132 in primary rat neurons after 24h and 72h, respectively, and 4 compounds significantly upregulated miR-132 in NGN2-iNs after 24h (Fig. 3A, Table S5). Notably, the cardiac glycosides, ouabain and digoxin, upregulated miR-132 in all conditions.
To investigate the dose response, we selected forskolin as the positive control, digoxin, ouabain, BIX02188, nitazoxanide, and pelitinib as the hits. We also included 6 additional cardiac glycosides (digitoxin, oleandrin, bufalin, bufotalin, cinobufagin, and proscillaridin A) and BIX02189, an analog of BIX02188. These compounds represent diverse chemical groups and mechanisms of action (Fig. 3B and Table S6). DIV14 primary rat cortical neurons were treated with doses ranging from 1 nM to 100 µM for 24h. Remarkably, all 8 cardiac glycosides upregulated miR-132 2.5-3-fold in the nM range, with proscillaridin A having the lowest EC50 of 3.2 nM (Table S6). Other compounds also dose-dependently upregulated miR-132 but with higher EC50. For all compounds tested, miR-212, which is co-transcribed and co-functional with miR-132 (29), was similarly upregulated at almost identical EC50, suggesting that the mechanism was largely transcriptional (Fig. S3A and Table S6). The cardiac glycosides proscillaridin A, oleandrin, digoxin, ouabain, and bufalin also upregulated miR-132 and miR-212 in a dose-dependent manner in human NGN2-iNs in the nM range (Fig. S3B, C and Table S6). However, BIX02188, which robustly upregulated miR-132 in primary rat neurons, had no effect on miR-132 in NGN2-iNs (Fig. S3B, C), suggesting potential differences between the two cell models.
To investigate the specificity of miR-132 upregulation by oleandrin and BIX02188, we measured the expression level of 10 other abundant neuronal miRNAs in rat primary cortical neurons after 24h treatment. When normalized to the geometric mean of all 12 miRNAs (30), only miR-132 and miR-212 were upregulated (Fig. S3D). The precursors pre-miR-132 and pre-miR-212 (Fig. 3C) were also upregulated by forskolin, BIX02118, and the cardiac glycosides, suggesting that these compounds activated the transcription of the miR-132/212 locus. Correspondingly, the upregulation of miR-132 by forskolin and oleandrin was completely blocked by pretreatment with the transcription inhibitor actinomycin D and partially blocked by the maximum tolerated doses of a CREB inhibitor (1 µM CREB-I) (Fig. 3D and S3E-H). As cardiac glycosides are conventional inhibitors of Na+/K+ pumps, we also knocked down ATP1A1 and ATP1A3, the dominant isoforms in neurons, with siRNAs. Knocking down either ATP1A1 or ATP1A3 also increased the expression of products of the miR-132/212 locus (Fig. 3E), suggesting that cardiac glycosides upregulated miR-132 by inhibiting their conventional targets.
Cardiac glycosides reduce miR-132 targets and protect against toxic insults in rodent neurons
To investigate the kinetics of miR-132 upregulation, we treated primary rat cortical neurons with 100 nM oleandrin and measured the expression of the precursor and the mature forms of miR-132 and miR-212 overtime (Fig. 4A, B). Both pre-miR-132 and pre-miR-212 were rapidly upregulated following treatment, peaked at 8h, and rapidly declined to baseline after 72h (Fig. 4A). Compared to their precursors, mature miR-132 and − 212 were upregulated at slower kinetics, peaked at 24h, then slowly declined but were still ~ 2-fold above baseline at 72h (Fig. 4B). We speculated that the increase in miR-132 expression would lead to the downregulation of its targets. Indeed, we observed a time-dependent downregulation of MAPT, FOXO3a, and EP300 mRNAs that matched the upregulation of miR-132 (Fig. 4C). mRNA targets were significantly reduced to ~ 50% of baseline at 24h and to ~ 75% of baseline at 72h, which was similar to the observed effects for miR-132 mimics 72h after transfection (Fig. S4A-C). Tau, pTau S202/T305 (AT8), pTau S396, and FOXO3a proteins were also downregulated, though the ratio of pTau: total Tau was unchanged (Fig. 4D-H). In primary PS19 mouse neurons that overexpress human mutant Tau-P301S (34), oleandrin upregulated miR-132 and downregulated both mouse MAPT and human MAPT after 72h treatment (Fig. S4D-H).
We further hypothesized that the upregulation of miR-132 by the cardiac glycosides would be neuroprotective against various disease-related stress insults, such as excitotoxic glutamate or Aβ oligomers (20). As several studies have reported possible neurotoxic effects associated with cardiac glycosides (35, 36), we first treated rat neurons at different ages in vitro (DIVs 7/14/21/28) with digoxin, oleandrin, and proscillaridin A for 96h before measuring cellular viability. Interestingly, DIV7 neurons were highly susceptible to cardiac glycoside toxicity, with significant loss of viability observed at the miR-132 EC100 for all compounds tested (Fig. 4I-K). However, mature neurons were more resistant to cardiac glycoside toxicity, and no loss of viability was observed at miR-132 EC100 for neurons treated at DIV14 or later (Fig. 4I-K).
To investigate neuroprotective effects, we first treated DIV21 rat neurons with oleandrin and proscillaridin A at EC100 for 24h, followed by 100 µM glutamate or 10 µM Aβ42. Proscillaridin A and oleandrin pretreatment rescued neuronal viability 72h after toxic insults without affecting viability at baseline (Fig. 4I, M). As we previously showed that miR-132 mimics rescued loss of viability in younger neurons treated with glutamate (20), we performed similar experiments in DIV7 neurons. We observed a slight loss of viability due to proscillaridin A at baseline (Fig. 4N). However, oleandrin and proscillaridin A rescued loss of viability caused by glutamate excitotoxicity (Fig. 4N). Oleandrin and proscillaridin A also partially and dose-dependently rescued neurite loss induced by glutamate without affecting neurite at baseline (Fig. 4O, S5).
Cardiac glycosides significantly reduce Tau and pTau in human iPSC-neurons
To investigate the effects of cardiac glycosides in human neurons, we utilized two additional iPSC-derived neural progenitor cell (NPC) lines: MGH-2046-RC1 derived from an individual with FTD carrying the autosomal dominant mutation Tau-P301L (P301L), and MGH-2069-RC1 derived from a healthy individual directly related to MGH-2046 (WT). When differentiated into neurons (iPSC-neurons) for 6–8 weeks, these NPCs represent well-established models for studying tauopathy phenotypes in patient-specific neuronal cells relative to a healthy control (37–39).
Since miR-132 regulates Tau metabolism (18) and Tau lowering is a promising therapeutic strategy for ADRD (37), we first investigated the effects of cardiac glycosides on Tau protein levels. All three tested cardiac glycosides strongly and dose-dependently downregulated Tau, as exemplified by proscillaridin A. The treatment led to a clear reduction in total Tau (TAU5 antibody) and pTau S396 in WT neurons and in P301L mutant neurons (Fig. 5A, D). For total Tau (TAU5), the upper band (> 50 kDa, monomeric Tau + post-translational modifications (PTMs)) was more intense at lower concentrations. With increasing concentrations, the upper band disappeared, whereas the lower band (< 50kDa, possibly non-pTau) became slightly more intense. This downward band shift suggested that proscillaridin A reduced both Tau accumulation and altered PTMs. Consistent with the latter, proscillaridin A reduced the monomeric form of pTau S396 (~ 50 kDa) as well as the high molecular weight oligomeric pTau (≥ 250 kDa)
RT-qPCR was performed on a matched set of WT and P301L iPSC-neurons and showed a dose-dependent reduction in MAPT mRNA, a large increase in pre-miR-132, and a smaller increase in mature miR-132 (Fig. 5B-C, E-F). Similar results were obtained with digoxin and oleandrin (Fig. S6). Further immunoblot results showed that in P301L iPSC-neurons, 72h treatment with 1 µM proscillaridin A, digoxin, or oleandrin reduced both soluble and insoluble total Tau and pTau S396 (Fig. S7A-C). The treatment also resulted in a dose-dependent reduction in miR-132 targets at the protein levels, including FOXO3a, EP300, GSK3β, and RBFOX1 (Fig. S7D-O).
For all compounds, the concentration of 10 µM was associated with > 70% reduction in Tau and pTau with 24h and 72h treatments. However, this concentration also reduced neuronal synaptic markers, including post-synaptic density protein 95 (PSD95), synapsin 1 (SYN1), and β-III-tubulin representative of microtubules’ structural integrity. These results suggest that cardiac glycosides can compromise neuronal integrity at high concentrations and with prolonged exposure. Nevertheless, for each compound, we observed a significant safety window in which Tau lowering was not associated with reduced synaptic or microtubule markers (Fig. 5G-R). In all graphs, the yellow shade indicates the dose range where the loss of at least 2 synaptic markers was 30% or less (Fig. 5G-R). Notably, WT neurons appeared more susceptible to loss of synaptic markers upon treatment than P301L neurons, particularly at 72h. For example, proscillaridin A was less toxic to P301L neurons than WT neurons (Fig. 5O-P, Q-R).
Cardiac glycosides are neuroprotective in human neuronal models of tauopathy
To examine the effects of the cardiac glycosides on neuronal viability, WT and P301L iPSC-neurons were treated with various doses of digoxin, oleandrin, and proscillaridin A for 24h or 72h. A dose-dependent loss of viability was observed with all three compounds, particularly at 72h. Tau-WT neurons had up to 30% loss of viability after 72h treatment, particularly at the highest dose of 10 µM (Fig. 6A-C). Interestingly, in Tau-P301L neurons, the toxicity observed was minimal, with < 10% viability loss at the highest concentrations at 72h (Fig. 6D-F). These results were consistent with the previous immunoblot data (Fig. 5G-R), showing that P301L neurons were more resistant to cardiac glycoside toxicity than WT neurons.
We next tested whether cardiac glycosides can protect human neurons from various stressors that specifically affect human iPSC-neurons expressing mutant Tau (39). These include the excitotoxic agonist of glutamatergic receptors NMDA, an inhibitor of the mitochondrial electron transport chain complex I, rotenone, and the aggregation-prone Aβ (1–42) amyloid peptide. Tau-P301L neurons differentiated for 8 weeks were pretreated with cardiac glycosides for 6h prior to adding stressors for 18h, and viability was measured at the 24h time point (Fig. 6G). Cardiac glycosides were added at 1 µM and 5 µM, which did not affect cell viability in P301L neurons at 24h (Fig. 6D-F). All cardiac glycosides significantly rescued neuronal viability in the presence of stressors (Fig. 8H-J). The rescue could also be observed with immunofluorescent staining (Fig. 6K). At baseline, 1 µM of digoxin, oleandrin, or proscillaridin A reduced Tau staining in agreement with the immunoblot data (Fig. 5D) without visibly affecting neuronal health. Treatment with the stressors led to a significant loss of neurites and cell bodies in neurons pretreated with vehicle alone, which was rescued by pretreatment with the cardiac glycosides. Overall, these results demonstrate that low concentrations of cardiac glycosides were neuroprotective in human tauopathy neurons.
Transcriptome analysis of human iPSC-neurons confirms shared pathways affected by cardiac glycosides
To further investigate the molecular mechanisms of cardiac glycosides’ neuroprotection, we profiled transcriptomes of human iPSC-neurons after 72h of treatment with increasing doses of digoxin, oleandrin, proscillaridin A or vehicle alone (0.1% DMSO) using RNA sequencing (Fig. 7A). Starting from low doses, cardiac glycosides remarkably changed the global transcriptomes of Tau-P301L neurons, as seen in principal component analysis (Fig. 7B), with a single principal component (PC1) being able to clearly separate controls from treatments. More importantly, three different cardiac glycosides regulated transcriptomes similarly and in a prominent dose-dependent manner (Fig. 7B). Differential expression analysis identified thousands of genes significantly regulated with fold-change higher than 4 (Fig. 7C). Many genes were related to neuronal health and activity, including the strongly upregulated ARC, which encapsulates RNA to mediate various forms of synaptic plasticity (40, 41), and downregulated MAPT and the SLITRK3/4/6 family, which plays a role in suppressing neurite outgrowth (42). We further focused on the biological pathways that were commonly regulated by all three cardiac glycoside compounds. Notably, these treatments affected many shared pathways (Fig. 7D). Downregulated genes belong to 74 pathways related to neuronal development, morphology, health, or activity (Fig. 7E). Upregulated genes were highly enriched in positive regulators of transcription, negative regulators of programmed cell death, and regulators of stress and unfolded protein response (Fig. 7F). Furthermore, dozens of transcription factors that had binding sites on MIR132 promoter and may upregulate its expression, including CREB5, were commonly upregulated by cardiac glycosides (Fig. 7G). The neuroprotective BDNF signaling pathway was significantly upregulated (Fig. S8). Therefore, while digoxin, oleandrin, and proscillaridin A all induced miR-132 expression, they likely regulated other pathways. Overall, shared transcriptomic alterations and regulated pathways further confirmed the common molecular mechanisms of action of cardiac glycosides and their ability to activate stress-protective programs in highly vulnerable Tau-mutant neurons (Fig. 7H).