SUMMARY
In polycystic kidney disease (PKD), microscopic tubules expand into macroscopic cysts. Among the world’s most common genetic disorders, PKD is inherited via heterozygous loss-of-function mutations, but theorized to require additional loss-of-function. To test this, we establish human pluripotent stem cells in allelic series representing four common nonsense mutations, using CRISPR base editing. When differentiated into kidney organoids, homozygous mutants spontaneously form cysts, whereas heterozygous mutants (original or base-corrected) express no phenotype. Using these, we identify eukaryotic ribosomal selective glycosides (ERSGs) as PKD therapeutics enabling ribosomal read-through of these same nonsense mutations. Two different ERSGs not only prevent cyst initiation, but also limit growth of pre-formed cysts, by partially restoring polycystin expression. Glycosides also accumulate in cyst epithelia in organoids and mice. Our findings define the human polycystin threshold as a surmountable drug target for pharmacological or gene therapy interventions, with relevance for understanding disease mechanism and future clinical trials.
INTRODUCTION
Human organoids and organ-on-chip devices are being developed to better understand disease mechanism and enable investigational new drug (IND) applications1. For genetic disorders, in which the human genome is a major therapeutic target, animal models are low throughput and may not fully recapitulate human conditions due to differences between species2. Simpler human cell cultures, such as monolayers, remain immature compared to tissues and lack heterocellularity and 3D architecture3. Human organoids and related systems provide a natural solution to these challenges, for a wide variety of disorders.
One such disorder is polycystic kidney disease (PKD), a severe, life-threatening genetic disease featuring progressive expansion of microscopic tubules into numerous fluid-filled cysts. Affecting approximately 1 in 1000 individuals worldwide, PKD is the leading monogenic cause of renal failure4,5. Tolvaptan, a vasopressin antagonist, can modestly slow cyst growth, but is associated with side effects of frequent thirst and risk of severe hepatotoxicity, preventing widespread usage in PKD patients6. Vasopressin activity in PKD is also primarily limited to the collecting ducts, whereas cysts also arise in other nephron segments7. Further, no treatments are available prior to cyst formation. Thus, there remains a large unmet clinical need for the development of new therapeutics for PKD.
PKD commonly results from loss-of-function mutations in PKD1 or PKD2, encoding polycystin-1 (PC1) and polycystin-2 (PC2), respectively8,9. Mutations are inherited in a germline heterozygous fashion, and somatic ‘second hit’ mutations are theorized to contribute to disease10–13. Thus, at the cellular level, PKD is proposed to involve a loss of polycystin function below heterozygosity, although this has been difficult to prove. Recent studies in mice suggest that restoring gene levels can be therapeutic in PKD, even after cysts form, but the relevance of this approach to humans is not clear, as mice are much less susceptible to PKD as heterozygotes14,15.
Nephron-like kidney organoids derived from human pluripotent stem cells (hPSCs) contain renal epithelial segments16–19, with great relevance for studying PKD. Organoids with knockout mutations in either PKD1 or PKD2 express a robust cystic phenotype not observed in isogenic controls17,20–22. Determining whether heterozygous organoids also show a cystic phenotype, similar to heterozygous patients, is important to provide insight into the fundamental mechanisms of PKD, and guide therapeutic strategies, such as gene correction. If repairing a single allele were sufficient to rescue the cystic phenotype, it would support future efforts towards monoallelic gene therapy. If, however, the cystic phenotype could not be rescued with a monoallelic edit, then it would indicate that both alleles need to be corrected for a gene therapy to take effect.
Another therapeutic strategy, which has yet to be tested, is to restore PC1 or PC2 expression using pharmacological agents that enable ribosomal read-through of in-frame nonsense mutations. Such mutations, which result in truncated proteins, constitute ~40% of PKD1 and ~50% of PKD2 cases23–28, and produce more severe disease, compared to non-truncating mutations26. Eukaryotic ribosomal selective glycosides (ERSGs) are aminoglycoside analogues, including the investigational drug ELX-02 (NB124) and the similarly active preclinical molecule ELX-10 (NB157)29,30. ERSGs permit eukaryotic ribosomal read-through of premature stop codons by stabilizing the “exo” conformation31–33, which allows a near-cognate tRNA to displace releasing factors34. ERSG activity is specific to premature stop codons as compared to nascent stop codons35. In preclinical evaluations for treatment of cystic fibrosis and cystinosis, ERSGs have demonstrated read-through and functional protein restoration both in vitro and in vivo36–38. In Phase 1 clinical trials, ELX-02 has been well tolerated with appropriate pharmacokinetics39,40. ELX-02 is preferentially absorbed by kidney tubular epithelial cells, which results in high exposure, but is optimized to avoid nephrotoxic side effects that arise with other aminoglycosides38.
The therapeutic potential of ERSGs for PKD has not previously been tested, because mouse and organoid models with in-frame nonsense mutations do not exist. CRISPR base editing is a newer technology that confers the potential to specifically introduce a single nucleotide change, instead of indels41,42, and can be more efficient and safer than homologous recombination following a double-strand break43,44. Here, we establish CRISPR base edited organoid models of PKD, with clinically relevant nonsense mutations amenable to targeting with ERSGs, to evaluate the potential of restoring polycystin dosage as a therapeutic approach.
RESULTS
Base Editing Generates PKD hPSCs with Nonsense Mutations
Four clinically documented PKD nonsense mutations were selected for targeting by CRISPR base editing in hPSCs: PKD1-R2430X, PKD1-Q3838X, PKD2-R186X, and PKD2-R872X (Fig. 1A and Fig. S1A)23. PKD1-R2430X and PKD2-R872X are the most common PKD patient nonsense truncating mutations in PKD1 and PKD2, respectively. The PKD1-R2430X site lies within a difficult-to-target region where six pseudogenes align, but we identified primers that enabled us to detect targeted editing events (Fig. S1B–C). The other two sites (PKD1-Q3838X and PKD2-R186X) were chosen in regions close in proximity to those for which we have previously established indel mutants modeling PKD17,20. PKD1-R2430 and PKD2-R872 sites were not conserved between mouse and human (Fig. S1D).
Fig. 1. CRISPR base editing generates hPSCs with PKD patient nonsense mutations.

A) Schematic of mutations selected for base editing: PKD1-R2430X, PKD1-Q3838X, PKD2-R186X, and PKD2-R872X. Antibody drawings indicate epitopes used in subsequent analyses. B) Schematic of CRISPR base editing workflow. CRISPR cytosine base editing (BIG-TREE method) produces BFP+ and GFP+ hPSCs transiently. GFP+ hPSCs isolated by FACS sorting are clonally expanded and screened. Illustrations are simplified and final cell lines no longer express GFP. C) Schematic summary of hPSC clones generated. Circles represent individual hPSC clones screened, color coded by genotype. Grey circles represent clones with additional edits at C bases within the guide sequence other than the targeted base. D) Representative chromatograms of C/C, C/T, and T/T clones for each targeted mutation. Dashed box indicates targeted base within guide sequence. E) Representative immunoblots of PC1 or PC2 in C/C, C/T, and T/T hPSC clones for each target mutation, with alpha-tubulin loading control. Carats (< symbols) indicate truncated protein bands. Dashed line indicates merging of lanes from same immunoblot. F) Quantification of full-length PC1 or full-length PC2 abundance relative to total PC1 or total PC2 abundance. n=3 independent clones per genotype; Welch’s ANOVA with Dunnett’s T3 multiple comparisons test. *p<.05, **p<.01, ***p<.001. Mean±SEM. See also Figure S1.
To generate multiple allelic series, we employed the Base-Edited Isogenic hPSC Line Generation Using a Transient Reporter for Editing Enrichment (BIG-TREE) method (Fig. 1B and Fig. S1E–F)45,46. Unedited C/C, heterozygous edited C/T, and homozygous edited T/T clones were identified for all four targeted PKD mutations (Fig. 1C–D)47. In total, 280 clones were screened for editing and multiple clones of each genotype were obtained (Fig. 1C–D and S1F–G). 31 null mutant clones and 77 heterozygous clones were generated in this process, in addition to non-mutant controls. Representative clones of each genotype were confirmed to exhibit features of normal hPSC morphology (Fig. S2A).
All four PKD targeted nonsense mutants lacked detectable expression of the targeted PC1 or PC2 full-length protein, respectively (Fig. 1E). PKD1-R2430X and PKD2-R872X clones expressed truncated forms of PC1 and PC2, respectively (Fig. 1E and S1H). For the carboxy-terminal Q3838X PC1 mutant, the truncated form could not be resolved using our antibody (7e12) against the amino terminus of PC1, which is cleaved from the carboxy terminus during PC1 maturation (Fig. 1E)48. However, levels of the PC1 amino terminus were reduced in the Q3838X mutant, suggesting degradation or nonsense-mediated decay (Fig. 1E–F). Another antibody suggested to recognize the C-terminus of human and mouse PC1 (E8–8C3C10) was unsuccessful in demonstrating specificity (Fig. S1H)48. In each of our allelic series, PC1 or PC2 protein levels reflected their genetic dosage, with no detectable expression in homozygous mutants, and ~50% expression in heterozygotes (Fig. 1E–F).
Kidney Organoids with Homozygous Nonsense Mutations Model PKD
To evaluate the capacity of nonsense mutant hPSCs to model PKD, we first differentiated them into nephron-like kidney organoids (Fig. 2A)17,49. Control and nonsense mutant kidney organoids expressed markers for podocytes, proximal tubules, and distal tubules in a similar fashion (Fig. 2B). Upon reaching maturity at day 18 of differentiation, kidney organoids were manually dissected out of adherent culture and transferred to suspension culture, a condition in which PKD organoids form cysts with high efficiency (Fig. 2A)20. 75–95% of PKD nonsense mutant kidney organoids formed cysts when maintained over fourteen days in suspension culture, > 15-fold higher compared to isogenic unedited controls that largely failed to produce cysts in side-by-side experiments (Fig. 2C–G and Fig. S2A). Rates of cystogenesis and cyst growth were similar across all four targeted PKD mutations (Fig. 2D–G). To account for potential clonal variability, we utilized multiple independent clones per genotype in all experiments. PKD kidney organoid cysts expressed diverse markers of proximal (LTL, CUBN, OAT1, SLC3A1) and distal (ECAD, MUC1, NKCC2, DBA) tubular regions of the nephron, which co-existed in cyst-lining epithelia while remaining partially compartmentalized (Fig. 2H–I). These findings supported the observation that PKD organoid cysts arise rapidly from large stretches of tubular epithelium comprising multiple nephron segments of origin20,22,50,51. Thus, base edited organoids with a spectrum of clinically-relevant mutations robustly recapitulated the PKD phenotype.
Fig. 2. Kidney organoids with homozygous nonsense mutations model PKD.

A) Schematic of PKD kidney organoid cystogenesis assay. B) Representative images of day 18 kidney organoids for isogenic unedited control and each targeted PKD mutant. Scale bars 50 μm. Lotus tetragonobolus lectin (LTL) labels proximal tubule cells, E-cadherin (ECAD) labels distal tubule cells, and podocalyxin (PODXL) labels podocytes. C) Representative images of kidney organoids at day 0 immediately following transfer to suspension culture and at day 14 following maintenance in suspension culture for isogenic unedited control and each targeted PKD mutant. Scale bar 500 μm. D) Quantification of the percentage of organoids forming cysts in suspension culture for paired PKD1 isogenic unedited C/C controls and homozygous edited T/T nonsense mutants. n=4 independent experiments per genotype. Mean±SEM. Some error bars too small to appear on graphs. Graphs show results from all four genotypes. Nonlinear regression extra-sum-of-squares F test p-values as indicated. E) Quantification of cyst area for paired PKD1 isogenic unedited C/C controls and homozygous edited T/T nonsense mutants. n=100 organoids pooled from 4 independent experiments per genotype. Mean±SEM. Some error bars too small to appear on graphs. Graphs show results from all four genotypes. F-G) Same as D-E but for PKD2 genotypes. Linear regression extra-sum-of-squares F test p-values as indicated. H) Representative confocal immunofluorescence images of PKD kidney organoid cysts stained with proximal and I) distal tubular markers: cubulin (CUBN), organic anion transport 1 (OAT1), solute carrier family 3 member 1 (SLC3A1), mucin 1 (MUC1), sodium-potassium chloride cotransporter 2 (NKCC2), and Dolichos biflorus agglutinin (DBA), with LTL and ECAD counterstains. n=4 organoids per condition representative of 3–4 independent biological replicates. Scale bar 50 μm. See also Figure S2 and Methods S1.
Heterozygosity Rescues PKD Cystogenesis
We assessed the phenotype of PKD heterozygous compared to homozygous mutant organoids, as well as isogenic controls, generated from our cohort of cytosine base edited hPSCs (Fig. 3A). In two allelic series, organoids heterozygous for either PKD1 or PKD2 nonsense truncations did not produce cysts at a significant rate, when compared with homozygous mutants, even after many weeks in culture (Fig. 3B–E and S3A). These findings indicated that the human gene dosage threshold to express PKD phenotype was below heterozygosity, i.e. ~50% of functional PC1 or PC2 relative to isogenic controls (above Fig. 1E–F).
Fig. 3. Heterozygosity rescues PKD cystogenesis.

A) Schematic of CRISPR cytosine base editing (C->T) outcomes for PKD nonsense mutations. B) Representative images of kidney organoids at day 14 following maintenance in suspension culture for isogenic unedited controls (C/C), heterozygous edited (C/T), and homozygous edited (T/T) mutants associated with PKD1-R2430X and PKD2-R186X genotypes. Scale bar 500 μm. C) Quantification of the percentage of organoids forming cysts in suspension culture for paired isogenic unedited C/C controls, heterozygous edited C/T, and homozygous edited T/T nonsense mutants. n=4 independent experiments per genotype. Mean±SEM. Nonlinear regression extra-sum-of-squares F test p-values as indicated. D) Quantification of cyst area for these samples. n=100 organoids pooled from 4 independent experiments per genotype. Mean±SEM. Linear regression extra-sum-of-squares F test p-values as indicated. E) Representative images of kidney organoids following 48 days in suspension culture. Scale bar 5 mm. F) Schematic of CRISPR adenine base editing (A->G) outcomes for PKD nonsense mutations. G) Schematic of CRISPR adenine base editing workflow. Starting pool of PKD2-R186X nonsense mutation cells have A/A at targeted base within guide sequence. CRISPR adenine base editing (XMAS-TREE method) produces mCherry+ and GFP+ hPSCs. GFP+ hPSCs isolated by FACS sorting are expanded and screened. H) Representative chromatograms of A/A and A/G clones. Dashed box indicates targeted base within guide sequence. I) Representative immunoblot of PC2 levels in A/A, A/G, and WT G/G clones with alpha-tubulin loading control. Dashed line indicates merging of lanes from same immunoblot. J) Quantification of PC2 abundance relative to alpha-tubulin loading control of A/A, A/G, and G/G clones. n=3 independent experiments with 2–4 A/A and A/G clones per genotype. Welch’s ANOVA with Dunnett’s T3 multiple comparisons test. **p<.01, ****p<.0001. Mean±SEM. K) Representative confocal immunofluorescence images of kidney organoids from A/A and A/G hPSCs. Scale bars 50 μm. L) Representative images of A/A and A/G kidney organoids at day 0 immediately following transfer to suspension culture and at days 14 and 30 following maintenance in suspension culture. Scale bar 500 μm for day 0 and 14. Scale bar 5 mm for day 30. M) Quantification of the percentage of A/A and A/G organoids forming cysts in suspension culture. n=4 independent experiments per genotype. Mean±SEM. Nonlinear regression extra-sum-of-squares F test p-value as indicated. N) Quantification of cyst area of A/A and A/G organoids in suspension culture. n=300 organoids pooled from 4 independent experiments per genotype. Mean±SEM. Linear regression extra-sum-of-squares F test p-value as indicated. Some error bars too small to appear on graphs. See also Figure S3 and Methods S1.
To determine if a genetic approach for correcting nonsense mutations could be a viable option for PKD gene therapy, we employed CRISPR adenine base editing to revert a mutated base to the consensus (non-mutant) sequence (Fig. 3F–G). We selected the PKD2-R186X site as it was the only mutation in our cohort that lied within an editing window of the DNA sequence amenable to adenine base editing without bystander effects (Fig. S3B). We utilized the Cas9-Mediated Adenosine Transient Reporter for Editing Enrichment (XMAS-TREE) method to edit hPSCs (Fig. 3G and Fig. S3C–D)52. By Sanger sequencing, unedited PKD2-R186X homozygous mutants (A/A) and heterozygous gene corrected (A/G) clones were identified (Fig. 3H)47. 76 clones were screened for editing and multiple heterozygous clones were obtained (Fig. S3E). Clones were maintained for many passages in culture and confirmed to exhibit features of normal hPSCs (Fig. S3F). Protein level changes were analyzed by immunoblot, where PKD2-R186X reversion rescued PC2 abundance to ~50% of non-mutant levels (Fig. 3I–J).
To evaluate the capacity of heterozygous corrected hPSCs to rescue PKD cystogenesis, we differentiated them into kidney organoids (Fig. 3K). To account for potential clonal variability, we utilized multiple independent clones per genotype. In side-by-side experiments, PKD2-R186X reversion reduced rates of cystogenesis from greater than 70% for unedited homozygous mutant organoids to ~ 10% for heterozygous gene corrected organoids, accompanied by a reduction in cross-sectional cyst area (Fig. 3L–N). In comparison to the original PKD2-R186X heterozygous organoids, the gene corrected PKD2-R186X heterozygous organoids appeared to form cysts at a slightly higher rate (Fig. 3B–C and Fig. 3L–N). This may reflect a batch effect between different experiments, or an off-target effect of repeated editing in these clones. Taken together, heterozygous organoids generated either from control (non-mutant) hPSCs by cytosine base editing, or from homozygous mutant hPSCs corrected by adenine base editing, demonstrated that mutations in a single copy of PKD1 or PKD2 were not sufficient to induce a cystic phenotype in human organoids.
We have previously shown that PC1 expression is dramatically reduced in hPSCs and organoids derived from PKD2−/− mutants with indel mutations20. Similarly, in our homozygous PKD2-R186X mutants generated using base editing, PC1 expression was greatly reduced (Fig. S3G–H). In gene-corrected heterozygous (PKD2+/−) cells, PC1 abundance was restored to nearly that of non-mutant levels (Fig. S3G–H). Thus the molecular phenotype of PC1 loss was a specific consequence of PC2 loss in human cells, and could be prevented by restoring a single copy of PC2.
Read-Through Treatments Safely Reduce PKD Cyst Formation
The innovation of nonsense mutation PKD organoids opened the potential to testing ERSG therapeutics that promote read-through. Two unique ERSGs, ELX-02 and ELX-10, were first tested in this model system using a ‘prophylactic treatment’ approach. Following terminal differentiation into nephron-like structures, but prior to PKD cyst formation, organoids were placed in individual wells to allow for consistent tracking over a 14-day treatment time course (Fig. 4A). We tested a dose range of 10 – 90 uM, which approximates systemic doses of ELX-02 that were previously shown to be well tolerated in humans subjects in vivo, and which correspond to the human equivalent doses from experiments conducted in cystinotic mice38–40. A dose-dependent decrease was observed in the percentage of organoids forming cysts across all four targeted PKD mutations in response to treatment with either ERSG compound (Fig. 4B–C and Fig. S4A). When treated with ERSGs, the percentage of organoids forming cysts dropped to lower than 25%, significantly lower than the 75% observed in untreated organoids (Fig. 4C). Moreover, we observed a dose-dependent decrease in cyst size, as measured by cross-sectional area, with both of these treatments and in all of our mutants (Fig. 4D). At low doses, a reduction in cyst area could still be detected even in cases where effect on the percentage of organoids was more modest (Fig. 4C–D).
Fig. 4. Prophylactic read-through treatment prevents PKD cyst formation.

A) Schematic of prophylactic read-through treatment workflow. Each independent trial consists of 10 organoids per condition as indicated in plate layouts. B) Representative images of organoids in each condition at treatment day 0 and 14 (brightfield and Calcein AM/propidium iodide stain). Scale bars 100 um. C) Quantification of the percentage of organoids forming cysts for each targeted PKD mutation in each treatment condition over 14 day treatment course. n=3 independent trials each with 10 organoids per condition. Mean±SEM. Some error bars too small to appear on graphs. Nonlinear regression extra-sum-of-squares F test p-values comparing all conditions indicated in upper left. For pairwise comparisons (Water vs 10uM, Water vs 30uM, Water vs 90uM), significance p<.05 indicated by orange asterisk next to respective dose at day 14. D) Quantification of cyst area for each targeted PKD mutation in each treatment condition over 14 day treatment course. n=30 organoids pooled from 3 independent trials each with 10 organoids per condition. Mean±SEM. Some error bars too small to appear on graphs. Linear regression extra-sum-of-squares F test p-values comparing all conditions indicated in upper left. For pairwise comparisons (Water vs 10uM, Water vs 30uM, Water vs 90uM), significance p<.05 indicated by orange asterisk next to respective dose at day 14. E) Quantification (violin plots) of propidium iodide fluorescence (dead) relative to Calcein AM fluorescence (live) for each targeted PKD mutation. n=30 organoids pooled from 3 independent trials each with 10 organoids per condition. F) Relative cytotoxicity compared to vehicle control as determined by LDH release assay following 7 days of treatment for each targeted PKD mutation. n=30 organoids pooled from 3 independent trials each with 10 organoids per condition. Welch’s ANOVA with Dunnett’s T3 multiple comparisons test. ns=not significant, *p<.05, **p<.01, ***p<.001, ****p<.0001. For all violin plots, solid line indicates median and dashed lines indicate quartiles. Curve fits and all p-values are provided in supplemental materials. See also Figure S4 and Methods S1.
To evaluate the safety of these compounds, we first tested a broad range of doses in organoids derived from one of our mutant cell lines (PKD1-R2430X) using a lactate dehydrogenase (LDH) release assay at day 7 of treatment and a live/dead (Calcein AM/propidium iodide) staining assay at the day 14 experimental end-point. Concentrations up to 100 uM of either ERSG did not induce significant toxicity, whereas toxicity was observed with 300 uM of either ERSG in both assays (Fig. S4B–D). The spread of LDH levels was higher for 100 μM ELX-02 than lower concentrations, suggesting a minor population of injured cells (Fig. S4D). By day 14, the LDH release assay no longer detected actively dying cells at toxic concentrations, reflecting the fact that these structures had died (Fig. S4E). Based on these findings, we then further analyzed toxicity within the 10–90 uM treatment window during our prophylactic efficacy testing with both ERSGs in all of the mutants. A slight increase in toxicity was detected at the 90 uM dose (Fig. 4B,E–F and Fig. S4F). However, this was subtle in comparison to the toxicity observed at the 300 uM dose in the initial screening we performed in the PKD1-R2430X line (Fig. S4B–E).
We next tested whether ERSGs could reduce the rate of growth for pre-established cysts using a ‘therapeutic treatment’ approach, in which organoids were allowed to develop cysts prior to treatment initiation (Fig. 5A). A dose-dependent decrease in cyst area fold change was observed across all four targeted PKD mutations in response to treatment with either ERSG compound (Fig. 5B–C and Fig. S4G). At the highest doses, on average, cyst growth plateaued following ERSG treatment (Fig. 5C). Interestingly, individual organoids occasionally showed a reduction in cyst size (reversal), compared to untreated organoids (Fig. 5D and Fig. S4H). For the therapeutic treatment assay, no substantial toxicity was detectable by either live/dead staining or LDH release assay (Fig. 5B,E–F and Fig. S4I). The reduced toxicity observed in these pre-established cysts, compared to pre-cystic structures (above Fig. 4), may reflect changes in physiology or metabolism arising during the process of cystogenesis. Collectively, these studies suggested a broadly beneficial effect of ERSGs on PKD cystogenesis, both at pre-cystic and post-cystic stages, without inducing significant cytotoxicity.
Fig. 5. Therapeutic read-through treatment slows PKD cyst expansion.

A) Schematic of therapeutic read-through treatment workflow. B) Representative images of organoids in each condition at treatment day 0 and 14 (brightfield and Calcein AM/propidium iodide stain). Scale bars 100 um. C) Quantification of the fold-change in cyst area relative to starting cyst area at day 0 for each targeted PKD mutation in each treatment condition over 14 day treatment course. n=30 organoids pooled from 3 independent trials each with 10 organoids per condition. Mean±SEM. Some error bars too small to appear on graphs. Nonlinear regression extra-sum-of-squares F test p-values comparing all conditions indicated in upper left. For pairwise comparisons (Water vs 10uM, Water vs 30uM, Water vs 90uM), significance p<.05 indicated by orange asterisk next to respective dose at day 14. D) Percentage of cysts with fold change less than 1 for each targeted PKD mutation in each treatment condition over 14 day treatment course. n=3 independent trials each with 10 organoids per condition. Mean±SEM. Some error bars too small to appear on graphs. Nonlinear regression extra-sum-of-squares F test p-values comparing all conditions indicated in upper left. For pairwise comparisons (Water vs 10uM, Water vs 30uM, Water vs 90uM), significance p<.05 indicated by orange asterisk next to respective dose at day 14. E) Quantification of propidium iodide fluorescence (dead) relative to Calcein AM fluorescence (live) for each targeted PKD mutation. n=30 organoids pooled from 3 independent trials each with 10 organoids per condition. F) Relative cytotoxicity compared to vehicle control as determined by LDH release assay following 7 days of treatment for each targeted PKD mutation. n=30 organoids pooled from 3 independent trials each with 10 organoids per condition. Welch’s ANOVA p-values as indicated in upper left. For Welch’s ANOVA where p<.05, Dunnett’s T3 multiple comparisons test pairwise significance is shown. ns=not significant, **p<.01. For all violin plots, solid line indicates median and dashed lines indicate quartiles. Curve fits and all p-values are provided in supplemental materials. See also Figure S4 and Methods S1.
Read-through Treatments are Specific for Nonsense Mutants
We sought to confirm that our efficacy findings were the result of on-target ERSG induced read-through by monitoring expression of full-length PC1 and PC2 in nonsense mutant kidney organoids following treatment with ERSGs. For these experiments, we focused only on doses up to 60 uM, since partial toxicity was observed at 90 uM in our system. PC1 levels were dramatically reduced in PKD2 mutants, compared to isogenic controls, reflecting a requirement for PC2 to stabilize PC1 (Fig. 6A–D)20. Across all PKD1 or PKD2 target mutation sites, there was a dose dependent increase in PC1 expression with ERSG treatment (Fig. 6A–D and S5A). At the 30 μM ELX-02 treatment concentration, this increase was ~50% of isogenic control levels (Fig. 6A–B).
Fig. 6. Read-through treatments are specific for nonsense mutants.

A) Representative immunoblots of PC1 with alpha-tubulin loading control from kidney organoids following 14 days of treatment. WT indicates untreated isogenic C/C control organoids. Carat (>) symbol at left indicates band of truncated protein. For PKD1-R2430X, blue arrowhead indicates full-length PC1 above truncated band. Dashed line indicates merging of lanes from same immunoblot. B) Quantification of immunoblot PC1 fold change following treatment with ELX-02 relative to WT PC1 abundance. n=3 independent biological replicates per genotype; Welch’s ANOVA with Dunnett’s T3 multiple comparisons test where *p<.05 and **p<.01. Welch’s ANOVA p-value as indicated for graphs without pairwise significance. Mean±SEM. C) Schematic depicting proposed effects of treatment with ELX compounds. D) Quantification of PC1 fold change following 10 day treatment with 30 uM ELX-10 relative to WT PC1 abundance for each targeted PKD mutation. n=3 independent biological replicates per genotype. Unpaired t-test with Welch’s correction where *p < .05. E) Representative images of PKD2−/− organoids with indicated frameshift mutation in each condition at day 0, 7, and 14 of treatment with water (vehicle), 10 uM, or 30 uM ELX-02 (blue) or ELX-10 (purple). Scale bar 100 um. F) Quantification of the percentage of organoids forming cysts for PKD2 frameshift mutant in each treatment condition over 14 day treatment course with ELX-02. n=3 independent trials each with 10 organoids per condition. Mean±SEM. Nonlinear regression extra-sum-of-squares F test p-values as indicated. G) Quantification of cyst area for PKD2 frameshift mutant over 14 day treatment course with ELX-02. n=30 organoids pooled from 3 independent trials each with 10 organoids per condition. Mean±SEM. Linear regression extra-sum-of-squares F test p-values as indicated. H-I) Same as F-G but for ELX-10 treatment. See also Figures S5–6.
Interestingly, in addition to an increase in full-length PC1, the PKD1-R2430X mutant also demonstrated accumulation of the truncated PC1 form (Fig. 6A and S5A). We hypothesized that this difference in truncated PC1 expression might reflect effects of ELX-02 on nonsense-mediated decay of the mRNA, which can be sequence dependent53. Indeed, we observed an increase in PKD1 mRNA levels for this R2430X mutant when treated with ELX-02, but not the other mutants (Fig. S5B).
We previously showed that PC1 is dramatically reduced in PKD2−/− hPSCs and organoids20. In our ERSG-treated PKD2 nonsense mutants, we observed restoration of PC1 to wild-type levels, suggesting that PC2 was being restored (Fig. 6A–C). PC2 has also been shown to promote the maturation of PC1 from a lower molecular weight, Endoglycosidase H (EndoH)-sensitive form in the endoplasmic reticulum (ER) to a higher molecular weight, EndoH-resistant form in post-ER membrane compartments54. Compared to untreated cells, the PC1 bands from the ERSG treatment groups in the PKD2 nonsense mutants showed an increase in both forms of PC1, but favored the lower molecular weight form, compared to wild type controls, suggesting that PC2 levels in these cells were not fully adequate to rescue PC1 trafficking and maturation. Read-through rates produced by aminoglycoside-derived compounds vary between proteins and mutation context, sometimes with low efficiency (< 5 %), which can pose a significant challenge for detection of read-through using endogenous protein levels55,56. Consistent with this, we were unable to directly detect changes in PKD2 transcript or PC2 protein abundance in our PKD2 mutants after treatment with ERSGs (Fig. S5C–E). This may reflect reduced sensitivity of reagents for detecting PC2 compared to PC1 or lower efficiency of read-through for the PKD2 mutation sites. Indeed, in our system, PC2 abundance was not readily detectable below 10–25% of that in control cells with no mutations (Fig. S5F–G). PC2 abundance is generally higher than PC1 abundance, thus a small quantity of PC2 may be sufficient to stabilize PC1 and contribute to function54.
To evaluate read-through capacity using an alternative method, we used a dual luciferase reporter assay in immortalized cells transfected with plasmids specific to each mutation57. A dose dependent increase was observed in the percentage of read-through by both ERSGs, on par or higher than read-through levels achieved by treatment with positive control compounds gentamicin and G418 (Fig. S5H). Consistent with our above observations, read-through efficiency was lower in the PKD2 mutants compared to PKD1 mutants. Read-through efficiency is context dependent on the nucleotide immediately downstream of the stop codon58,59. Both of the PKD1 mutants have a cytosine base and both of the PKD2 mutants have a guanine base at this position, likely explaining the observed differences in read-through efficiency in these cohorts. For the PKD2 mutations, read-through levels plateaued at 3–6%, lower than the detection threshold of our protein abundance assay (Fig. S5F–H). Thus, the increase in PC1, which is stabilized by PC2, in ERSG-treated PKD2 nonsense mutants was consistent with the hypothesis that low levels of PC2 were being expressed54,60.
Certain small molecules, such as mefloquine, can function as enhancers of aminoglycoside-mediated read-through61,62. Mefloquine is thought to potentiate the effects of read-through compounds by binding to the eukaryotic 80S ribosome, inducing a favorable conformational change61,63. We tested whether mefloquine addition might enable us to detect direct effects of ERSGs on PKD2 transcript levels. In undifferentiated hPSCs, PKD2 levels were significantly reduced in PKD2-R186X mutant hPSCs versus isogenic controls, likely reflecting a process of nonsense-mediated decay in these cells (Fig. S5I). Treatment with ELX-02 resulted in a slight increase in transcript, which was enhanced ~40 % by supplementation of ELX-02 with mefloquine, but not mefloquine alone (Fig. S5I). Levels of PKD1 in these cells were unaffected by mefloquine, suggesting that this was effect was specific to the mutant PKD2 transcript (Fig. S5J). These findings further supported a direct effect of ERSGs on PKD2.
To test whether the ERSG treatment response in our organoid model was specific to nonsense mutations, we applied ELX-02 or ELX-10 to organoids derived from PKD2−/− hPSCs with homozygous deletion mutations (p.Ser88SerfsTer207) generated with CRISPR-Cas920. As such mutations are not amenable to read-through, there was no effect on cystogenesis in PKD2−/− organoids lacking nonsense mutations when treated with either ERSG at doses up to 30 uM (Fig. 6E–I and Fig. S6A–D). To further control for off-target effects of ERSGs, we added ELX-02 to organoid cultures at an earlier stage of differentiation, and observed that that renal vesicle structures maintained in 10 uM or 30 uM progressed normally to terminal differentiation (Fig. S6E–F). Taken together, the observed restoration of full-length PC1 protein and specificity of the treatment response to nonsense mutations indicate on-target activity of ERSGs in our organoid model system.
ERSGs Accumulate in Kidney Cysts
Aminoglycosides are taken up by nephron epithelia, but whether this extends to PKD cysts is unclear38,64–67. As ELX drugs with fluorescent conjugates are not available, we instead utilized Texas red-gentamicin (GTTR), a fluorophore-conjugated aminoglycoside that closely resembles the structure of ELX-02 and ELX-10, and is known to accumulate in kidney tubules in vivo in a similar manner38,64–66. In PKD organoid cysts, GTTR uptake into the cyst lining epithelial cells occurred over a period of hours (Fig. 7A). In fixed samples, GTTR signal was present in both proximal (LTLhiECADlo) and distal (ECADhiLTLlo) cells in an intracellular localization pattern (Fig. 7B and Fig. S7A).
Fig. 7. Aminoglycosides localize to PKD cysts.

A) Representative time course images of PKD organoid cysts treated with Gentamicin-Texas Red (GTTR) or water (vehicle). Red arrow indicates start of treatment. n=4 organoids from 3 independent differentiations. Scale bar 100 um. B) Representative confocal 20X and 40X images of PKD organoid cysts treated with 30 uM GTTR followed by fixation at 48 hours post treatment initiation. LTL stains proximal tubule and ECAD stains distal tubule. Scale bars 100 um. C) Schematic depicting GTTR uptake assay. D) Representative confocal images of cysts receiving PBS or GTTR perfusion. Two representative cysts are shown for GTTR, with digital zoom below. Scale bar 200 um PBS and GTTR, 50 um for zoom. E) Line scan analysis of PKD cysts after perfusion (mean ± SEM, n = 20 cysts per condition pooled from 4 male Pkd1RC/RC mice of C57BL/6J background). Yellow arrows in (D) represent direction of line scans. See also Figure S7.
In mouse studies in vivo, GTTR was previously shown to persist in proximal and distal tubules for 2–3 days after administration65,66. Similarly, in PKD organoids, GTTR administered for 2 hours and subsequently removed (washout) was still visible in cyst lining epithelium 48 hours later, and co-localized with markers of proximal and distal tubules (Fig. S7B–C). When cysts were treated with both GTTR and unlabeled gentamicin as a competitor for binding, the fluorescent signal decreased in a dose-dependent manner with increasing concentrations of the unlabeled compound, demonstrating specificity (Fig. S7D–E). Thus, aminoglycoside derivatives accumulated to a steady-state level inside organoid cyst-lining epithelial cells.
To test accumulation in vivo, we injected GTTR into the vasculature and immediately harvested the kidneys and processed them unfixed for fluorescence analysis (Fig. 7C)51. This was performed using Pkd1RC/RC mice, which are homozygous for a hypomorphic mutation to Pkd1, and develop progressive PKD68,69. Analysis of tissue sections revealed that a subset of cyst-lining epithelia readily accumulated GTTR (Fig. 7D–E). Immunofluorescence analysis on serial tissue sections indicated that GTTR accumulated in cysts lacking specific markers of collecting ducts (AQP2+) or proximal tubules (LTL+), whereas cysts that stained positive for AQP2 did not exhibit significant uptake in these short-term GTTR injections (Fig. S7F–G). Cysts lacking specific markers develop progressively in these PKD mice, and are thought to represent dedifferentiation events in which a subset of cysts lose their segment markers of origin68,69. Uptake of GTTR was also observed in non-cystic proximal tubules, as expected from prior analyses, which further supported the specificity of the signal observed in these experiments (Fig. S7F–G)38,64–67. Thus, ERSGs were capable of uptake into PKD cysts in vivo.
DISCUSSION
Nonsense Mutants Test Therapeutic Approaches
Conceptually, the major advance of the hPSC-derived PKD organoid system over other cellular models is that it demonstrates a clear genotype-phenotype relationship specific to PKD. Our comprehensive cohort of base edited allelic series, representing > 130 sequenced clonal cell lines, and gene corrected lines, robustly supports this conclusion and our foundational studies in this area.
In therapeutic ‘trials in a dish’ on pre-established cysts, we found that ERSGs significantly limited cyst expansion. This expands the potential to treat PKD patients with advanced disease in order to further delay disease progression. A recent report shows that induced expression of PKD2 from a BAC transgene reversed cysts beginning at 13 weeks of age in an in vivo mouse model of PKD15. Our own data also indicate a dose-dependent level of cyst reversal in organoids treated with read-through ERSGs, although we were only able to detect this in individual organoids and not at the level of the general population, using a compound to induce treatment effects rather than a purely genetic strategy. Notably, even a modest increase in PC1 abundance was sufficient to reduce cystogenesis.
The ERSGs used in this study were selectively optimized to reduce nephrotoxicity compared to traditional aminoglycoside antibiotics31. As cytotoxicity can potentially masquerade as efficacy in cystogenesis experiments, it is important to conduct experiments with multiple doses and appropriate controls. For experiments in which treatment was initiated prior to cyst formation, we observed partial toxicity at 90 μM, possibly because the tubules are more functional prior to cyst formation and accumulate higher doses of drug. Thus, to prevent cyst initiation, doses of 30 μM or below would be more suitable, to avoid off-target toxicity.
Support for the Two-Hit Hypothesis
Organoids with heterozygous nonsense mutations, lacking only one copy of either PKD1 or PKD2, failed to produce robust cysts. This was not due to compensatory upregulation of the mutant allele, as we confirmed that expression of the mutant protein was only ~50 % in our heterozygotes. Rather, this finding supports a threshold model in which additional events lower the functional dosage of PC1 or PC2 below heterozygosity, enabling cystogenesis to occur10–13. We also demonstrate that heterozygous restoration of PKD2 expression by CRISPR base editing significantly reduces the rate of cystogenesis, establishing a ‘floor’ of ~50 % protein level that is sufficient to rescue the phenotype, and demonstration that base editing can constitute a viable therapeutic strategy for PKD.
Other groups recently described kidney organoids developed using the conventional CRISPR-Cas9 system (indel generating) or PKD patient-derived, in which an increased level of cystogenesis was observed in heterozygotes compared to wild type, although this remained much less than isogenic homozygotes70,71. In those studies, which utilized a distinct differentiation protocol, cystogenesis in PKD organoids (heterozygous or homozygous) required the addition of forskolin, a strong artificial secretagogue that can also trigger cysts in non-PKD structures20,22,72. In contrast, PKD organoids generated using our differentiation protocol form cysts robustly without the use of forskolin, a more natural context. Thus, in light of our findings, the data in human organoids support the conclusion that heterozygotes resemble control organoids, rather than homozygotes.
In hPSCs and organoids with indel mutations, PC2 loss results in a dramatic reduction in full-length PC1 protein20. Interestingly, heterozygous PKD2+/− hPSCs show a near-complete rescue of PC1 expression, demonstrating that full dosage of PC2 is not required. Similarly, treatment of PKD2−/− nonsense mutants with read-through compounds greatly increases PC1, even though PC2 levels are only partially rescued. This dosage-independent effect of PC2 on PC1 levels in human cells appears to be distinct from mice, in which Pkd2 exhibits a dosage-dependent effect on PC1 maturation54.
Limitations of the Study
The nephron-like organoids used in these studies lack collecting ducts. Distinct protocols are needed to differentiate hPSCs into the ureteric lineage that produces collecting ducts, and currently these are neither fully mature nor capable of modeling PKD cystogenesis without forskolin70,73. Our work reveals that aminoglycoside derivatives can reach kidney cysts in vivo, but as the injections we performed were short in duration, further studies are needed to demonstrate biodistribution, safety, and efficacy in longer-term treatments of animals. Proximal tubules are a logical segment to target for therapy, given their tendency to take up aminoglycosides. Achieving efficacy in distal tubules and collecting ducts would further enhance the clinical relevance of this approach but may require higher doses of drug.
Read-through compounds are typically capable of achieving only relatively modest efficiency, which varies depending on the read-through target55,56. Future studies may employ more advanced and sensitive techniques, such as mass spectrometry, to quantify the PC1 and PC2 polypeptide fragments generated by read-through. Given the very robust effects of these read-through compounds on PC1 levels, compared to relatively minor effects on PC2, the complete molecular mechanisms remain elusive, and it is possible that the compounds may contribute specifically to PC1 stabilization via additional mechanisms, which remain to be determined. This would be an interesting topic for future studies, which may lead to targets for PC1 restoration. For the gene therapy aspect, we have been able to achieve rescue by gene correction, but it would be valuable to establish techniques to efficiently modify mature organoids, which would be the equivalent of the kidneys in patients. Our toolkit of genetic and chemical tools might be further developed to assess such questions in future studies.
STAR★METHODS
RESOURCE AVAILABILITY
Lead Contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Benjamin Freedman (benof@uw.edu).
Materials Availability
All unique reagents generated in the study are available from the lead contact under material transfer request.
Data and Code Availability
All data reported in this paper will be shared by the lead contact upon request. This paper does not report original code. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request. Scans of full-length Western blots are available on Mendeley Data (doi:10.17632/8t4y3f8zs8.1) at the following link: https://data.mendeley.com/preview/8t4y3f8zs8?a=a9a27a05-74fc-450e-8ff6-7eeebbd4fcae
EXPERIMENTAL MODELS AND STUDY PARTICIPANT DETAILS
Animals
All animal studies were conducted in accordance with all relevant ethical regulations under protocols approved by the Institutional Animal Care and Use Committee at the University of Washington in Seattle. C57BL/6J Pkd1RC/RC mice (MGI: 5476822), males ≥ 18 months old were utilized for the experiments. Mice were maintained on a standard diet under standard pathogen-free housing conditions, with food and water freely available.
Cell Lines
WTC11 hPSCs (Coriell 25256, female) were maintained on 1% reduced growth factor GelTrex (Life Technologies) in mTeSR1 media (Stem Cell Technologies) and dissociated with Accutase (Stem Cell Technologies). HeLa cells (CCL-2, ATCC, female) were cultured in DMEM (Thermo Fisher), supplemented with 10% of non-heat-inactivated fetal bovine serum (Sigma-Aldrich) and 1% PenStrep (Thermo Fisher).
METHOD DETAILS
Kidney Organoid Differentiation.
hPSCs were dissociated with Accutase (Stem Cell Technologies) and seeded at a density of 1500 cells per well in a 24-well plate pre-coated with GelTrex in mTeSR1 supplemented with 10 μM Rho-kinase inhibitor Y27632 (Tocris Bioscience). The media was replaced with 500 μL mTeSR1 + 1:40 growth factor reduced Matrigel (Corning) at 16 hours, 500 μL mTeSR1 at 36 hours, Advanced RPMI + Glutamax (Life Technologies) + 12 μM CHIR99021 (Tocris Bioscience) at 50 hours, and RB - Advanced RPMI + 1x Glutamax + 1x B27 Supplement (Life Technologies) at 86 hours49. RB was changed two days later and every three days thereafter. Organoids were differentiated for 18 days from the time of plating.
For cystogenesis assays, adherent organoids were microdissected with a 23-gauge syringe needle from 24-well plates on an inverted phase-contrast microscope, and carefully transferred using a P1000 pipette into an ultra-low attachment 6-well plate (Corning) containing 2 mL RB. RB media was changed by gravity every three days. Wells were imaged using a Nikon TiE inverted widefield microscope. Cyst percentage and area were measured using FIJI software. Kidney organoid cysts were identified as having a bright, translucent thin-walled epithelium with 3D curvature surrounding a hollow, fluid-filled space. This is in contrast to the kidney organoid body that was identified as having a dull, dense multilayer architecture occasionally associated with a stromal cell layer. Cross-sectional cyst area was traced using the free-form tool in FIJI, outlining only the areas of translucency while excluding areas identified as the organoid body.
CRISPR Base Editing.
Gene alignments and amino acid alignments were performed in Benchling. Base editing guide sequences (listed in Supplemental Table 1) were generated using the Benchling CRISPR Design and Analyze Guides tool with base editing selected as the design type. Guides were optimized for highest base editing scores as predicted from sequence context and target C position41 as well as lowest number of off-target sites. BIG-TREE cytosine base editing plasmids were obtained from Addgene (pEF-AncBE4max #138270, pDT-sgRNA #138271, and pEF-BFP #138272)45. Guide oligonucleotides with CACC (sense) and AAAC (antisense) 5’-overhangs were duplexed and cloned into the pDT-sgRNA plasmid at the BbsI cut site using a Quick Ligation Kit (New England BioLabs). All intermediate plasmid products were confirmed via Sanger sequencing (Genewiz). 500,000 WTC11 hPSCs (Coriell 25256) were seeded in a 6 well plate with mTeSR1 media supplemented with 10 μM Rho-kinase inhibitor Y27632 24 hours prior to transfection. pEF-AncBE4max (1800 ng), pDT-sgRNA with unique guide (900 ng), and pEF-BFP (900 ng) were co-transfected using 8 μL Lipofectamine Stem transfection reagent (Themo Fisher). Media was replaced with mTeSR1 24 hours post transfection. 48 hours post transfection, hPSCs were dissociated as single cells using Accutase and passed through a 35 μm cell strainer. BFP+ and GFP+ cells were isolated using a FACS Aria cell sorter and plated at low density in recovery media (mTeSR1 supplemented with 1X CloneR (Stem Cell Technologies)). Recovery media was changed 48 hours post plating and supplemented with 20% recovery media by volume 72 hours post plating without a full change. Media was changed to standard mTeSR1 96 hours post plating followed by daily changes until small colonies formed. Individual colonies were manually isolated with a 10 μl pipet and transferred to a 96-well plate mTeSR1 media supplemented with 10 μM Rho-kinase inhibitor Y27632 followed by daily changes with mTeSR1 media until confluent. Clones were then passaged with one replica plate dissociated and temporarily frozen in FreSR-S (Stem Cell Technologies) at −80°C and the other replica plate used for genotype screening. Genomic DNA was prepared from the resulting clonal cell lines using QuickExtract DNA extraction solution (Lucigen). The targeted region was PCR amplified using Platinum Hot Start 2X Master Mix (Thermo Fisher) by thermocycling at 94°C or 2 minutes followed by 35 cycles of 94°C, 57°C, and 72°C for 30 seconds each followed by 72°Cfor 5 minutes. PCR products sizes were confirmed on a 1% agarose gel and were purified using ExoSAP-IT (Applied Biosystems) prior to Sanger sequencing (Genewiz). Genomic DNA primer sequences are listed in Supplemental Table 1. Resultant sequencing files were analyzed with EditR software for chromatogram alignment and detection of editing47. All isogenic sets for a targeted mutation site were then thawed in parallel from temporary storage at −80°C and expanded for long-term storage in liquid nitrogen. Isogenic control clones were obtained through the same CRISPR base editing procedure but were found to have no modification at the target site upon screening.
For reversion of the PKD2-R186X mutation, the guide sequence (listed in Supplemental Table 1) was generated using the CRISPR RGEN Tools BEdesigner optimized for location of target base within the editing window sequence and lowest number of off-target binding sites74. XMAS-TREE adenine base editing plasmids were obtained from Addgene pEF-ABEmax (#164415), pDT-sgRNA-XMAS-1x (#164413), and pEF-XMAS-1xStop (#164411)52. The same protocol was followed as described above for adenine base editing except cell sorting was performed for mCherry+ and GFP+ cells. All oligonucleotides and PCR primers were synthesized by Integrated DNA Technologies.
Read-Through Compound Treatment.
For prophylactic treatment assays, organoids were micro dissected as described above. Single organoids were then transferred to individual wells of an ultra-low attachment round bottom 96 well plate (Corning) using a P200 pipette. Organoids were maintained in 100 μL RB media supplemented with water, ELX-02 (NB124), or ELX-10 (NB157) at indicated concentrations. On days 3, 5, 7, 10, and 12, RB media was changed by gravity with removal of 50 μL existing media followed by addition of 50 μL fresh media supplemented with water, ELX-02, or ELX-10 at 2x concentration. Therapeutic treatment assays were performed in the same way, but starting with cystic organoids that had been maintained in suspension for 7 days prior to transfer. For LDH release assays, 25 μL of media per well was transferred to a white 96 well plate (Cellstar). LDH-Glo cytotoxicity assay was performed according to manufacturer’s protocol (Promega). Gentamicin-Texas Red (GTTR) organoid experiments were performed in the same manner with removal of 50 μL existing media followed by addition of 50 μL fresh media supplemented with GTTR (AAT Bioquest 24300), with or without unconjugated gentamicin (Gibco 15710–064). Washout was performed by 3 successive changes of normal media.
Immunoblotting.
Undifferentiated hPSCs or derived organoids were maintained in adherent culture to obtain sufficient material for lysis. The same dosing schedule was used for organoid treatments as described above – day 0, 3, 5, 7, 10, and 12. Once hPSCs were confluent or on day 10 (ELX-10)/day 14 (ELX-02) of organoid treatment, cells were lysed using RIPA buffer (Thermo Fisher) with complete mini EDTA-free protease inhibitor (Sigma), PhosSTOP (Sigma), and benzonase nuclease (Sigma). Samples were centrifuged at 13,000 rpm for 10 minutes at 4°C to remove cell debris. Resulting lysates were quantified using a BCA protein assay kit (Thermo Fisher). Samples were prepared with 50 μg protein in Laemmli buffer (Bio-Rad) containing 2-mercaptoethanol (Sigma). Precision Plus Protein Kaleidoscope Standards (Bio-Rad) were loaded as a marker for molecular weights. Samples were separated in a 4–20% gel (Bio-Rad) and transferred onto a PVDF membrane. The PVDF membrane was blocked with 5% non-fat milk and probed with primary antibodies. The antibodies used were anti-PC1 7e12 (sc-130544; Santa Cruz), anti-PC1 E8 (E8–8C3C10, Kerafast), anti-PC2 (sc-25749; Santa Cruz), anti-αtubulin (2144s; Cell Signaling), anti-rabbit HRP (1706515; Bio-Rad), and anti-mouse HRP (176516; Bio-Rad). Signal was captured using Pierce ECL Western Blotting Substrate (ThermoFisher Scientific) and ProSignal ECL Blotting Film (Prometheus). Band intensities were quantified with FIJI Gel Analyzer and normalized to α-tubulin loading control.
Immunostaining and Imaging.
Kidney organoids or mouse tissue slices were fixed in 4% paraformaldehyde (Electron Microscopy Sciences) in PBS for 15 minutes at room temperature. Samples were washed with PBS, blocked in 5% donkey serum (Millipore)/0.3% Triton-X-100/PBS for 1 hour, and incubated overnight in 3% bovine serum albumin/PBS with primary antibodies. The following day, samples were washed with PBS and incubated overnight with Alexa-Fluor secondary antibodies (Invitrogen) and DAPI (Thermo Scientific). Prior to imaging, samples were washed with PBS and subsequently imaged in PBS. Primary antibodies used were biotinylated LTL (FL-1320, Vector Labs), DBA (Vector Labs RL-1032), ECAD (Abcam ab11512), PODXL (R&DAF1658), CUBN (Invitrogen PA5–83684), SLC3A1 (Sigma HPA038360), OAT1 (Invitrogen PA5–26244), MUC1 (Abcam ab15481), NKCC2 (Proteintech 18970–1-AP), and AQP2 (Abcam 199975). Calcein AM (Invitrogen) and Propidium Iodide (Thermo Fisher) stains were performed according to manufacturer’s instructions. Confocal fluorescence images were captured using a Yokogawa W1 Spinning Disk confocal head mounted on an inverted Nikon Ti widefield microscope. For cystogenesis assays, wells were imaged using a Nikon TiE inverted widefield microscope. For compound treatment assays, brightfield images were captured using the transmitted light detector on a Leica SP8 microscope. Cyst percentage and area were quantified using FIJI. Dead/live ratios were quantified as mean fluorescent intensity measurements on max intensity projections of organoids which were traced manually using FIJI. For suspension days 30 and 48, organoids were imaged overhead using an iPhone 12.
RT-PCR.
RNA was prepared using the Quick RNA MiniPrep kit (Zymo). 666 ng RNA was reverse transcribed using the SuperScriptIV Reverse Transcription kit (Invitrogen). For each independent experiment, quantitative RT–PCR reactions were run in triplicate using cDNA (diluted 1:2, 300 nM primers, and PowerUp SYBR Green Master Mix (Applied Biosystems). For read-through enhancer treatments, confluent hPSCs were treated with ELX-02 with or without mefloquine hydrochloride (Milipore Sigma M2319). For these experiments, 1000ng RNA was reverse transcribed and the cDNA was diluted 1:300. qPCR primer sequences are listed in Supplemental Table 1. mRNA fold change was calculated using 2−μμCT with actin as the housekeeping gene.
Dual Luciferase Assay.
HeLa cells were plated in a 96 well format at a density of 7500 cells per well in DMEM (Thermo Fisher), supplemented with 10% of non-heat-inactivated fetal bovine serum (Sigma-Aldrich). Cellular transfection occurred one day later using 0.1 μL Turbofect (ThermoFisher) and 25 ng DNA (pSGDluc plasmid with insert sequences as listed in Supplemental Table 1 (GenScript)). Compound treatment was initiated 24 h post transfection. FF and RNL luciferase measurements were made 24 h after compound treatment using the Dual Luciferase kit (Promega) as per the manufacturer protocol. Briefly, cells were lysed with 25 μL of Passive Lysis Buffer. 10 μL of lysate was transferred to a secondary plate and 25 μL of Luciferase Assay Reagent II was added. FF activity was measured on an Envision plate reader (PerkinElmer). 25 μL of Stop & Glo reagent was then added to each well followed by RNL activity measurements in the Envision plate reader. For each well, raw FF were normalized to raw RNL to obtain FF/RNL ratios. FF/RNL ratios of mutant plasmids were then normalized to wild-type plasmids. The percent increase in read-through activity was derived by calculating the difference between vehicle and compound-induced read-through, then dividing that value by the read-through value for vehicle and expressing the result as a percentage.
In Vivo Perfusion Experiments.
Pkd1RC/RC mice (≥ 18 months old) were sacrificed through isoflurane overdose and immediately incised through the chest and nicked at the vena cava with a 27-gauge needle. Keeping pressure on the vena cava, mice were perfused systemically through the heart with a syringe containing 10 ml of PBS, followed by a second syringe containing either 2.5 ml of PBS (control) or 2.5 ml of PBS + 83.33 μM Gentamicin-Texas Red (AAT Bioquest 24300). Kidneys were excised immediately and embedded directly in OCT without fixation or sucrose equilibration. 20 μm thick kidney sections were cut using a cryostat, mounted on a microscope slide in OCT, and imaged on a confocal microscope with 10X objective.
QUANTIFICATION AND STATISTICAL ANALYSIS
Statistical analyses were performed using GraphPad Prism Software. Information regarding specific statistical tests used, representation of data in graphs, and replicates for each experiment are included in the corresponding figure legend. For nonlinear regression, p-values were calculated with an extra sum-of-squares F test using an exponential plateau model comparing YM and k values for percentage of organoids forming cysts and percentage of cysts with fold change less than 1, a straight-line model comparing slopes for cyst area, and a logistic growth model comparing YM and k values for cyst fold change. Nonlinear regression curves for all plots are provided in Data S2-S3. At least 2 unique clones per genotype were used in each experiment. In total, over 11,500 images of individual organoids were analyzed.
Supplementary Material
KEY RESOURCES TABLE (KRT).
| REAGENT or RESOURCE | SOURCE | IDENTIFIER |
|---|---|---|
| Antibodies | ||
| anti-PC1 7e12 | Santa Cruz | sc-130554 |
| anti-PC1 E8 | Kerafast | E8-8C3C10 |
| anti-PC2 H280 | Santa Cruz | sc-25749 |
| anti-α-tubulin | Cell Signaling | 2144 |
| anti-ECAD | Abcam | ab11512 |
| anti-CUBN | Invitrogen | PA5-83684 |
| anti-OAT1 | Invitrogen | PA5-26244 |
| anti-MUC1 | Abcam | ab15481 |
| anti-NKCC2 | Proteintech | 18970-1-AP |
| anti-SLC3A1 | Sigma | HPA038360 |
| anti-PODXL | R&D | AF1658 |
| anti-AQP2 | Abcam | ab199975 |
| Bacterial and Virus Strains | ||
| NEB Stable Compotent E. Coli | New England Biolabs | C3040H |
| Chemicals, Peptides, and Recombinant Proteins | ||
| Calcein AM | Invitrogen | C3099 |
| Propodium Iodide | Invitrogen | P1304MP |
| CHIR99021 | Tocris | 4423 |
| ELX-02 | Eloxx Pharmaceuticals | NB142 |
| ELX-10 | Eloxx Pharmaceuticals | NB157 |
| Biotinylated LTL | Vector Labs | FL-1320 |
| DBA | Vector Labs | RL-1032 |
| DAPI | Cayman Chemical | 14285 |
| Gentamicin | Gibco | 15710-064 |
| GTTR | AAT Bioquest | 24300 |
| Mefloquine | Milipore Sigma | M2319 |
| Critical Commercial Assays | ||
| Quick Ligation Kit | New England BioLabs | M2200 |
| Lipofectamine Stem Transfection Reagent | Invitrogen | STEM00001 |
| Sanger Sequencing | Genewiz | https://www.genewiz.com |
| QuickExtract | Lucigen | QE09050 |
| Platinum Hot Start PCR Master Mix | Invitrogen | 13000012 |
| ExoSAP-IT Express | Applied Biosystems | 75001 |
| LDH-Glo Cytotoxicity Assay | Promega | J2381 |
| Quick RNA MiniPrep Kit | Zymo | R1054 |
| SuperScript IV | Invitrogen | 18091050 |
| PowerUp SYBR Green Master Mix | Applied Biosystems | A25743 |
| TurboFect | Thermo Fisher | R0531 |
| Dual-Glo Luciferase Assay | Promega | E2920 |
| Experimental Models: Cell Lines | ||
| WTC11 | Coriell | 25256 |
| HeLa | ATCC | CCL-2 |
| Experimental Models: Organisms/Strains | ||
| Mouse: C57BL/6J Pkd1RC/RC | The Jackson Laboratory | MGI:5476822 |
| Oligonucleotides | ||
| Cytosine Base Editing gRNAs | This Paper: Supplemental Table 1 | N/A |
| Genomic DNA Sequencing Primers | This Paper: Supplemental Table 1 | N/A |
| qPCR Primers | This Paper: Supplemental Table 1 | N/A |
| Dual Luciferase Insert Sequences | This Paper: Supplemental Table 1 | N/A |
| Recombinant DNA | ||
| pEF-AncBE4max | Addgene | #138270 |
| pDT-sgRNA | Addgene | #138271 |
| pEF-BFP | Addgene | #138272 |
| pEF-ABEmax | Addgene | #164415 |
| pDT-sgRNA-XMAS-1x | Addgene | #164413 |
| pEF-XMAS-1xStop | Addgene | #164411 |
| pSGDluc | Addgene | #87323 |
| Software and Algorithms | ||
| EditR | Kluesner, M.G., Nedveck, D.A., Lahr, W.S., Garbe, J.R., Abrahante, J.E., Webber, B.R., and Moriarity, B.S. (2018). EditR: A Method to Quantify Base Editing from Sanger Sequencing. Cris. J. 1, 239–250. 10.1089/crispr.2018.0014. | https://moriaritylab.shinyapps.io/editr_v10/ |
| CRISPR Guide RNA Design Tool | Benchling | https://www.benchling.com/crispr |
| Fiji (2.0.0) | Schindelin, J., Arganda-Carreras, I., Frise, E., Kaynig, V., Longair, M., Pietzsch, T., … Cardona, A. (2012). Fiji: an open-source platform for biological-image analysis. Nature Methods, 9(7), 676–682. 10.1038/nmeth.2019 | https://imagej.net/software/fiji/ |
| Prism (9.3.1) | GraphPad | https://www.graphpad.com |
| BioRender | BioRender | https://www.biorender.com |
| CRISPR RGEN Tools BEdesigner | Hwang, GH., Park, J., Lim, K. et al. Web-based design and analysis tools for CRISPR base editing. BMC Bioinformatics 19, 542 (2018). https://doi.org/10.1186/s12859-018-2585-4 | http://www.rgenome.net/be-designer/ |
ACKNOWLEDGEMENTS
We thank Vijay Modur, Vasudeo Badarinarayana, Megan Cox, and Ali Hariri (Eloxx Pharmaceuticals), Jonathan Himmelfarb (JH) Isabella Jennings (UW), and Scott Medina (SM, University of Pittsburgh) for technical support and helpful discussions. Illustrations were created with BioRender.com. Studies were supported by an Eloxx Pharmaceuticals Award, NIH Awards R01DK117914 (BSF), UH3TR002158 (JH), UH3TR003288 (JH), U01DK127553 (BSF), U01AI176460 (BSF), U2CTR004867 (BSF), UC2DK126006 (BSF), P30DK089507 (pilot to BSF), R21DK128638 (SM), R35GM142902 (SM), the Lara Nowak-Macklin Research Fund, and a Washington Research Foundation fellowship (CT).
Footnotes
DECLARATION OF INTERESTS
Eloxx Pharmaceuticals is developing ELX-02 and ELX-10 as investigational drugs, and holds patents related to their use. BSF is an inventor on patents and/or patent applications related to human kidney organoid differentiation and modeling of PKD in this system. BSF holds ownership interest in Plurexa LLC.
REFERENCES
- 1.Rumsey JW, Lorance C, Jackson M, Sasserath T, McAleer CW, Long CJ, Goswami A, Russo MA, Raja SM, Gable KL, et al. (2022). Classical Complement Pathway Inhibition in a “Human-On-A-Chip” Model of Autoimmune Demyelinating Neuropathies. Adv. Ther. 5, 2200030. 10.1002/adtp.202200030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Vandamme TF (2014). Use of rodents as models of human diseases. J. Pharm. Bioallied Sci. 6, 2–9. 10.4103/0975-7406.124301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Li M, and Izpisua Belmonte JC (2019). Organoids - Preclinical Models of Human Disease. N. Engl. J. Med. 380, 569–579. 10.1056/NEJMra1806175. [DOI] [PubMed] [Google Scholar]
- 4.DALGAARD OZ (1957). Bilateral polycystic disease of the kidneys; a follow-up of 284 patients and their families. Dan. Med. Bull. 4, 128–133. [PubMed] [Google Scholar]
- 5.Aung TT, Bhandari SK, Chen Q, Malik FT, Willey CJ, Reynolds K, Jacobsen SJ, and Sim JJ (2021). Autosomal Dominant Polycystic Kidney Disease Prevalence among a Racially Diverse United States Population, 2002 through 2018. Kidney360 2, 2010–2015. 10.34067/KID.0004522021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Torres VE, Chapman AB, Devuyst O, Gansevoort RT, Grantham JJ, Higashihara E, Perrone RD, Krasa HB, Ouyang J, and Czerwiec FS (2012). Tolvaptan in patients with autosomal dominant polycystic kidney disease. N. Engl. J. Med. 367, 2407–2418. 10.1056/NEJMoa1205511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gattone VH 2nd, Wang X, Harris PC, and Torres VE (2003). Inhibition of renal cystic disease development and progression by a vasopressin V2 receptor antagonist. Nat. Med. 9, 1323–1326. 10.1038/nm935. [DOI] [PubMed] [Google Scholar]
- 8.The European Polycystic Kidney Disease Consortium (1994). The polycystic kidney disease 1 gene encodes a 14 kb transcript and lies within a duplicated region on chromosome 16. Cell 77, 881–894. 10.1016/0092-8674(94)90137-6. [DOI] [PubMed] [Google Scholar]
- 9.Mochizuki T, Wu G, Hayashi T, Xenophontos SL, Veldhuisen B, Saris JJ, Reynolds DM, Cai Y, Gabow PA, Pierides A, et al. (1996). PKD2 a Gene for Polycystic Kidney Disease That Encodes an Integral Membrane Protein. Science (80-. ). 272, 1339 LP – 1342. 10.1126/science.272.5266.1339. [DOI] [PubMed] [Google Scholar]
- 10.Qian F, Watnick TJ, Onuchic LF, and Germino GG (1996). The Molecular Basis of Focal Cyst Formation in Human Autosomal Dominant Polycystic Kidney Disease Type I. Cell 87, 979–987. 10.1016/S0092-8674(00)81793-6. [DOI] [PubMed] [Google Scholar]
- 11.Watnick T, He N, Wang K, Liang Y, Parfrey P, Hefferton D, St George-Hyslop P, Germino G, and Pei Y (2000). Mutations of PKD1 in ADPKD2 cysts suggest a pathogenic effect of trans-heterozygous mutations. Nat. Genet. 25, 143–144. 10.1038/75981. [DOI] [PubMed] [Google Scholar]
- 12.Wu G, D’Agati V, Cai Y, Markowitz G, Park JH, Reynolds DM, Maeda Y, Le TC, Hou HJ, Kucherlapati R, et al. (1998). Somatic inactivation of Pkd2 results in polycystic kidney disease. Cell 93, 177–188. 10.1016/s0092-8674(00)81570-6. [DOI] [PubMed] [Google Scholar]
- 13.Wu G, Tian X, Nishimura S, Markowitz GS, D’Agati V, Park JH, Yao L, Li L, Geng L, Zhao H, et al. (2002). Trans-heterozygous Pkd1 and Pkd2 mutations modify expression of polycystic kidney disease. Hum. Mol. Genet. 11, 1845–1854. 10.1093/hmg/11.16.1845. [DOI] [PubMed] [Google Scholar]
- 14.Lakhia R, Ramalingam H, Chang C-M, Cobo-Stark P, Biggers L, Flaten A, Alvarez J, Valencia T, Wallace DP, Lee EC, et al. (2022). PKD1 and PKD2 mRNA cis-inhibition drives polycystic kidney disease progression. Nat. Commun. 13, 4765. 10.1038/s41467-022-32543-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Dong K, Zhang C, Tian X, Coman D, Hyder F, Ma M, and Somlo S (2021). Renal plasticity revealed through reversal of polycystic kidney disease in mice. Nat. Genet. 10.1038/s41588-021-00946-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Taguchi A, Kaku Y, Ohmori T, Sharmin S, Ogawa M, Sasaki H, and Nishinakamura R (2014). Redefining the in vivo origin of metanephric nephron progenitors enables generation of complex kidney structures from pluripotent stem cells. Cell Stem Cell 14, 53–67. 10.1016/j.stem.2013.11.010. [DOI] [PubMed] [Google Scholar]
- 17.Freedman BS, Brooks CR, Lam AQ, Fu H, Morizane R, Agrawal V, Saad AF, Li MK, Hughes MR, Werff R Vander, et al. (2015). Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nat. Commun. 6, 8715. 10.1038/ncomms9715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Morizane R, Lam AQ, Freedman BS, Kishi S, Valerius MT, and Bonventre JV (2015). Nephron organoids derived from human pluripotent stem cells model kidney development and injury. Nat. Biotechnol. 33, 1193–1200. 10.1038/nbt.3392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Takasato M, Er PX, Chiu HS, Maier B, Baillie GJ, Ferguson C, Parton RG, Wolvetang EJ, Roost MS, Chuva de Sousa Lopes SM, et al. (2015). Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature 526, 564–568. 10.1038/nature15695. [DOI] [PubMed] [Google Scholar]
- 20.Cruz NM, Song X, Czerniecki SM, Gulieva RE, Churchill AJ, Kim YK, Winston K, Tran LM, Diaz MA, Fu H, et al. (2017). Organoid cystogenesis reveals a critical role of microenvironment in human polycystic kidney disease. Nat. Mater. 16, 1112–1119. 10.1038/nmat4994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wilson PD (2008). Mouse models of polycystic kidney disease. Curr. Top. Dev. Biol. 84, 311–350. 10.1016/S0070-2153(08)00606-6. [DOI] [PubMed] [Google Scholar]
- 22.Tran T, Song CJ, Nguyen T, Cheng S-Y, McMahon JA, Yang R, Guo Q, Der B, Lindström NO, Lin DC-H, et al. (2022). A scalable organoid model of human autosomal dominant polycystic kidney disease for disease mechanism and drug discovery. Cell Stem Cell 29, 1083–1101.e7. 10.1016/j.stem.2022.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.PKD Foundation ADPKD Variant Database. https://pkdb.mayo.edu/variants.
- 24.Rossetti S, Hopp K, Sikkink RA, Sundsbak JL, Lee YK, Kubly V, Eckloff BW, Ward CJ, Winearls CG, Torres VE, et al. (2012). Identification of gene mutations in autosomal dominant polycystic kidney disease through targeted resequencing. J. Am. Soc. Nephrol. 23, 915–933. 10.1681/ASN.2011101032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Rossetti S, Strmecki L, Gamble V, Burton S, Sneddon V, Peral B, Roy S, Bakkaloglu A, Komel R, Winearls CG, et al. (2001). Mutation analysis of the entire PKD1 gene: genetic and diagnostic implications. Am. J. Hum. Genet. 68, 46–63. 10.1086/316939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Cornec-Le Gall E, Audrézet M-P, Chen J-M, Hourmant M, Morin M-P, Perrichot R, Charasse C, Whebe B, Renaudineau E, Jousset P, et al. (2013). Type of PKD1 mutation influences renal outcome in ADPKD. J. Am. Soc. Nephrol. 24, 1006–1013. 10.1681/ASN.2012070650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Rossetti S, Consugar MB, Chapman AB, Torres VE, Guay-Woodford LM, Grantham JJ, Bennett WM, Meyers CM, Walker DL, Bae K, et al. (2007). Comprehensive molecular diagnostics in autosomal dominant polycystic kidney disease. J. Am. Soc. Nephrol. 18, 2143–2160. 10.1681/ASN.2006121387. [DOI] [PubMed] [Google Scholar]
- 28.Garcia-Gonzalez MA, Jones JG, Allen SK, Palatucci CM, Batish SD, Seltzer WK, Lan Z, Allen E, Qian F, Lens XM, et al. (2007). Evaluating the clinical utility of a molecular genetic test for polycystic kidney disease. Mol. Genet. Metab. 92, 160–167. 10.1016/j.ymgme.2007.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kandasamy J, Atia-Glikin D, Shulman E, Shapira K, Shavit M, Belakhov V, and Baasov T (2012). Increased Selectivity toward Cytoplasmic versus Mitochondrial Ribosome Confers Improved Efficiency of Synthetic Aminoglycosides in Fixing Damaged Genes: A Strategy for Treatment of Genetic Diseases Caused by Nonsense Mutations. J. Med. Chem. 55, 10630–10643. 10.1021/jm3012992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Sabbavarapu NM, Shavit M, Degani Y, Smolkin B, Belakhov V, and Baasov T (2016). Design of Novel Aminoglycoside Derivatives with Enhanced Suppression of Diseases-Causing Nonsense Mutations. ACS Med. Chem. Lett. 7, 418–423. 10.1021/acsmedchemlett.6b00006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Shulman E, Belakhov V, Wei G, Kendall A, Meyron-Holtz EG, Ben-Shachar D, Schacht J, and Baasov T (2014). Designer aminoglycosides that selectively inhibit cytoplasmic rather than mitochondrial ribosomes show decreased ototoxicity: a strategy for the treatment of genetic diseases. J. Biol. Chem. 289, 2318–2330. 10.1074/jbc.M113.533588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Irina P, B. AR, Muminjon D, P. SJ, Alexandre U, Angelica F, Tom CC-W, Marat Y, C. BS, and Gulnara Y (2017). Aminoglycoside interactions and impacts on the eukaryotic ribosome. Proc. Natl. Acad. Sci. 114, E10899–E10908. 10.1073/pnas.1715501114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Burke JF, and Mogg AE (1985). Suppression of a nonsense mutation in mammalian cells in vivo by the aminoglycoside anthiotics G–418 and paromomycin. Nucleic Acids Res. 13, 6265–6272. 10.1093/nar/13.17.6265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Fourmy D, Yoshizawa S, and Puglisi JD (1998). Paromomycin binding induces a local conformational change in the A-site of 16 S rRNA. J. Mol. Biol. 277, 333–345. 10.1006/jmbi.1997.1551. [DOI] [PubMed] [Google Scholar]
- 35.Crawford D, Alroy I, Sharpe N, Goddeeris M, and Williams G (2020). ELX-02 generates protein via premature stop codon read-through without inducing native stop codon read-through proteins. J. Pharmacol. Exp. Ther, jpet.120.265595. 10.1124/jpet.120.265595. [DOI] [PubMed] [Google Scholar]
- 36.Crawford DK, Mullenders J, Pott J, Boj SF, Landskroner-Eiger S, and Goddeeris MM (2021). Targeting G542X CFTR nonsense alleles with ELX-02 restores CFTR function in human-derived intestinal organoids. J. Cyst. Fibros. 10.1016/j.jcf.2021.01.009. [DOI] [PubMed] [Google Scholar]
- 37.Xue X, Mutyam V, Tang L, Biswas S, Du M, Jackson LA, Dai Y, Belakhov V, Shalev M, Chen F, et al. (2014). Synthetic aminoglycosides efficiently suppress cystic fibrosis transmembrane conductance regulator nonsense mutations and are enhanced by ivacaftor. Am. J. Respir. Cell Mol. Biol. 50, 805–816. 10.1165/rcmb.2013-0282OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Brasell EJ, Chu LL, Akpa MM, Eshkar-Oren I, Alroy I, Corsini R, Gilfix BM, Yamanaka Y, Huertas P, and Goodyer P (2019). The novel aminoglycoside, ELX-02, permits CTNSW138X translational read-through and restores lysosomal cystine efflux in cystinosis. PLoS One 14, e0223954–e0223954. 10.1371/journal.pone.0223954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Leubitz A, Frydman-Marom A, Sharpe N, van Duzer J, Campbell KCM, and Vanhoutte F (2019). Safety, Tolerability, and Pharmacokinetics of Single Ascending Doses of ELX-02, a Potential Treatment for Genetic Disorders Caused by Nonsense Mutations, in Healthy Volunteers. Clin. Pharmacol. Drug Dev. 8, 984–994. 10.1002/cpdd.647. [DOI] [PubMed] [Google Scholar]
- 40.Leubitz A, Vanhoutte F, Hu M, Porter K, Gordon E, Tencer K, Campbell K, Banks K, and Haverty T (2021). A Randomized, Double-Blind, Placebo-Controlled, Multiple Dose Escalation Study to Evaluate the Safety and Pharmacokinetics of ELX-02 in Healthy Subjects. Clin. Pharmacol. Drug Dev. n/a. 10.1002/cpdd.914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Komor AC, Kim YB, Packer MS, Zuris JA, and Liu DR (2016). Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420–424. 10.1038/nature17946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Koblan LW, Doman JL, Wilson C, Levy JM, Tay T, Newby GA, Maianti JP, Raguram A, and Liu DR (2018). Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction. Nat. Biotechnol. 36, 843–848. 10.1038/nbt.4172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zeng J, Wu Y, Ren C, Bonanno J, Shen AH, Shea D, Gehrke JM, Clement K, Luk K, Yao Q, et al. (2020). Therapeutic base editing of human hematopoietic stem cells. Nat. Med. 26, 535–541. 10.1038/s41591-020-0790-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Frangoul H, Altshuler D, Cappellini MD, Chen Y-S, Domm J, Eustace BK, Foell J, de la Fuente J, Grupp S, Handgretinger R, et al. (2021). CRISPR-Cas9 Gene Editing for Sickle Cell Disease and β-Thalassemia. N. Engl. J. Med. 384, 252–260. 10.1056/NEJMoa2031054. [DOI] [PubMed] [Google Scholar]
- 45.Brookhouser N, Tekel SJ, Standage-Beier K, Nguyen T, Schwarz G, Wang X, and Brafman DA (2020). BIG-TREE: Base-Edited Isogenic hPSC Line Generation Using a Transient Reporter for Editing Enrichment. Stem Cell Reports 14, 184–191. 10.1016/j.stemcr.2019.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Standage-Beier K, Tekel SJ, Brookhouser N, Schwarz G, Nguyen T, Wang X, and Brafman DA (2019). A transient reporter for editing enrichment (TREE) in human cells. Nucleic Acids Res. 47, e120. 10.1093/nar/gkz713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kluesner MG, Nedveck DA, Lahr WS, Garbe JR, Abrahante JE, Webber BR, and Moriarity BS (2018). EditR: A Method to Quantify Base Editing from Sanger Sequencing. Cris. J. 1, 239–250. 10.1089/crispr.2018.0014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Yu S, Hackmann K, Gao J, He X, Piontek K, García-González MA, Menezes LF, Xu H, Germino GG, Zuo J, et al. (2007). Essential role of cleavage of Polycystin-1 at G protein-coupled receptor proteolytic site for kidney tubular structure. Proc. Natl. Acad. Sci. U. S. A. 104, 18688–18693. 10.1073/pnas.0708217104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cruz NM, and Freedman BS (2019). Chapter 7 - Differentiation of human kidney organoids from pluripotent stem cells. In Methods in Kidney Cell Biology - Part A, T. B. T.-M. in Weimbs CB, ed. (Academic Press; ), pp. 133–150. 10.1016/bs.mcb.2019.05.003. [DOI] [PubMed] [Google Scholar]
- 50.Czerniecki SM, Cruz NM, Harder JL, Menon R, Annis J, Otto EA, Gulieva RE, Islas LV, Kim YK, Tran LM, et al. (2018). High-Throughput Screening Enhances Kidney Organoid Differentiation from Human Pluripotent Stem Cells and Enables Automated Multidimensional Phenotyping. Cell Stem Cell 22, 929–940.e4. 10.1016/j.stem.2018.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Li SR, Gulieva RE, Helms L, Cruz NM, Vincent T, Fu H, Himmelfarb J, and Freedman BS (2022). Glucose absorption drives cystogenesis in a human organoid-on-chip model of polycystic kidney disease. Nat. Commun. 13, 7918. 10.1038/s41467-022-35537-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Brookhouser N, Nguyen T, Tekel SJ, Standage-Beier K, Wang X, and Brafman DA (2020). A Cas9-mediated adenosine transient reporter enables enrichment of ABE-targeted cells. BMC Biol. 18, 1–15. 10.1186/s12915-020-00929-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Nagy E, and Maquat LE (1998). A rule for termination-codon position within intron-containing genes: when nonsense affects RNA abundance. Trends Biochem. Sci. 23, 198–199. 10.1016/s0968-0004(98)01208-0. [DOI] [PubMed] [Google Scholar]
- 54.Gainullin VG, Hopp K, Ward CJ, Hommerding CJ, and Harris PC (2015). Polycystin-1 maturation requires polycystin-2 in a dose-dependent manner. J. Clin. Invest. 125, 607–620. 10.1172/JCI76972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Floquet C, Hatin I, Rousset J-P, and Bidou L (2012). Statistical analysis of readthrough levels for nonsense mutations in mammalian cells reveals a major determinant of response to gentamicin. PLoS Genet. 8, e1002608. 10.1371/journal.pgen.1002608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Bidou L, Hatin I, Perez N, Allamand V, Panthier J-J, and Rousset J-P (2004). Premature stop codons involved in muscular dystrophies show a broad spectrum of readthrough efficiencies in response to gentamicin treatment. Gene Ther. 11, 619–627. 10.1038/sj.gt.3302211. [DOI] [PubMed] [Google Scholar]
- 57.Grentzmann G, Ingram JA, Kelly PJ, Gesteland RF, and Atkins JF (1998). A dual-luciferase reporter system for studying recoding signals. RNA 4, 479–486. [PMC free article] [PubMed] [Google Scholar]
- 58.Manuvakhova M, Keeling K, and Bedwell DM (2000). Aminoglycoside antibiotics mediate context-dependent suppression of termination codons in a mammalian translation system. RNA 6, 1044–1055. 10.1017/s1355838200000716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Pranke IM, Varilh J, Hatton A, Faucon C, Girodon E, Dreano E, Chevalier B, Karri S, Reix P, Durieu I, et al. (2022). The U UGA C sequence provides a favorable context to ELX-02 induced CFTR readthrough. J. Cyst. Fibros. 10.1016/j.jcf.2022.10.010. [DOI] [PubMed] [Google Scholar]
- 60.Cai Y, Fedeles SV, Dong K, Anyatonwu G, Onoe T, Mitobe M, Gao J-D, Okuhara D, Tian X, Gallagher A-R, et al. (2014). Altered trafficking and stability of polycystins underlie polycystic kidney disease. J. Clin. Invest. 124, 5129–5144. 10.1172/JCI67273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Ferguson MW, Gerak CAN, Chow CCT, Rastelli EJ, Elmore KE, Stahl F, Hosseini-Farahabadi S, Baradaran-Heravi A, Coltart DM, and Roberge M (2019). The antimalarial drug mefloquine enhances TP53 premature termination codon readthrough by aminoglycoside G418. PLoS One 14, e0216423. 10.1371/journal.pone.0216423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Omachi K, Kai H, Roberge M, and Miner JH (2022). NanoLuc reporters identify COL4A5 nonsense mutations susceptible to drug-induced stop codon readthrough. iScience 25, 103891. 10.1016/j.isci.2022.103891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wong W, Bai X-C, Sleebs BE, Triglia T, Brown A, Thompson JK, Jackson KE, Hanssen E, Marapana DS, Fernandez IS, et al. (2017). Mefloquine targets the Plasmodium falciparum 80S ribosome to inhibit protein synthesis. Nat. Microbiol. 2, 17031. 10.1038/nmicrobiol.2017.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Sandoval RM, Bacallao RL, Dunn KW, Leiser JD, and Molitoris BA (2002). Nucleotide depletion increases trafficking of gentamicin to the Golgi complex in LLC-PK1 cells. Am. J. Physiol. Renal Physiol. 283, F1422–9. 10.1152/ajprenal.00095.2002. [DOI] [PubMed] [Google Scholar]
- 65.Wang Q, Kachelmeier A, and Steyger PS (2010). Competitive antagonism of fluorescent gentamicin uptake in the cochlea. Hear. Res. 268, 250–259. 10.1016/j.heares.2010.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Bellairs JA, Redila VA, Wu P, Tong L, Webster A, Simon JA, Rubel EW, and Raible DW (2022). An in vivo Biomarker to Characterize Ototoxic Compounds and Novel Protective Therapeutics. Front. Mol. Neurosci. 15, 944846. 10.3389/fnmol.2022.944846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Muther RS, and Bennett WM (1981). Cyst fluid antibiotic concentrations in polycystic kidney disease: differences between proximal and distal cysts. Kidney Int. 20, 519–522. 10.1038/ki.1981.170. [DOI] [PubMed] [Google Scholar]
- 68.Hopp K, Ward CJ, Hommerding CJ, Nasr SH, Tuan H-F, Gainullin VG, Rossetti S, Torres VE, and Harris PC (2012). Functional polycystin-1 dosage governs autosomal dominant polycystic kidney disease severity. J. Clin. Invest. 122, 4257–4273. 10.1172/JCI64313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Hopp K, Hommerding CJ, Wang X, Ye H, Harris PC, and Torres VE (2015). Tolvaptan plus pasireotide shows enhanced efficacy in a PKD1 model. J. Am. Soc. Nephrol. 26, 39–47. 10.1681/ASN.2013121312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kuraoka S, Tanigawa S, Taguchi A, Hotta A, Nakazato H, Osafune K, Kobayashi A, and Nishinakamura R (2020). PKD1-dependent renal cystogenesis in human induced pluripotent stem cell-derived ureteric bud/collecting duct organoids. J. Am. Soc. Nephrol. 31, 2355–2371. 10.1681/ASN.2020030378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Shimizu T, Mae S-I, Araoka T, Okita K, Hotta A, Yamagata K, and Osafune K (2020). A novel ADPKD model using kidney organoids derived from disease-specific human iPSCs. Biochem. Biophys. Res. Commun. 529, 1186–1194. 10.1016/j.bbrc.2020.06.141. [DOI] [PubMed] [Google Scholar]
- 72.Low JH, Li P, Chew EGY, Zhou B, Suzuki K, Zhang T, Lian MM, Liu M, Aizawa E, Rodriguez Esteban C, et al. (2019). Generation of Human PSC-Derived Kidney Organoids with Patterned Nephron Segments and a De Novo Vascular Network. Cell Stem Cell 25, 373–387.e9. 10.1016/j.stem.2019.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Taguchi A, and Nishinakamura R (2017). Higher-Order Kidney Organogenesis from Pluripotent Stem Cells. Cell Stem Cell 21, 730–746.e6. 10.1016/j.stem.2017.10.011. [DOI] [PubMed] [Google Scholar]
- 74.Gaudelli NM, Komor AC, Rees HA, Packer MS, Badran AH, Bryson DI, and Liu DR (2017). Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551, 464–471. 10.1038/nature24644. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All data reported in this paper will be shared by the lead contact upon request. This paper does not report original code. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request. Scans of full-length Western blots are available on Mendeley Data (doi:10.17632/8t4y3f8zs8.1) at the following link: https://data.mendeley.com/preview/8t4y3f8zs8?a=a9a27a05-74fc-450e-8ff6-7eeebbd4fcae
