Abstract β-propeller protein-associated neurodegeneration (BPAN) is a rare X-linked neurodegenerative disorder caused by mutations in the WDR45 gene, yet its molecular mechanisms remain poorly understood. Here, we identify a role for WDR45 in stress granule (SG) disassembly, mediated through its phase separation with Caprin-1. We demonstrate that WDR45 forms gel-like condensates via its WD5 domain, which competitively displaces G3BP1 from Caprin-1 to promote SG disassembly. BPAN-associated WDR45 mutations impair condensate formation and Caprin-1 interaction, leading to delayed SG disassembly, which correlates with earlier disease onset. WDR45 depletion also exacerbates amyotrophic lateral sclerosis-associated pathological SGs, highlighting its broader relevance to neurodegenerative diseases. Using iPSC-derived midbrain neurons from a BPAN patient, we demonstrate delayed SG recovery, directly linking WDR45 dysfunction to neurodegeneration. These findings establish WDR45 as a critical regulator of SG dynamics, uncover a potential molecular basis of BPAN pathogenesis, and identify therapeutic targets for neurodegenerative diseases associated with SG dysregulation. Subject terms: Mechanisms of disease, RNA metabolism, Neurodegenerative diseases, Intrinsically disordered proteins __________________________________________________________________ BPAN is a rare neurodegenerative disease caused by WDR45 mutations. Here, the authors discover that WDR45 can competitively displace G3BP1 from Caprin-1 to promote stress granule disassembly, a function that is disrupted by BPAN-associated WDR45 mutations. Introduction β-propeller protein-associated neurodegeneration (BPAN) is a rare X-linked neurodegenerative disorder characterized by developmental delays in childhood and progressive neurodegeneration in adulthood^[56]1,[57]2. Neuroimaging studies of BPAN patients have shown abnormal iron deposition in the basal ganglia, particularly in the globus pallidus, putamen, and substantia nigra^[58]3. In BPAN, degeneration of dopaminergic neurons and iron deposition in the substantia nigra pars compacta are implicated in the development of bradykinesia, a hallmark feature resembling Parkinson’s disease^[59]3,[60]4. Mutations in the gene encoding the WD repeat domain 45 (WDR45) protein have been identified as a primary cause of BPAN^[61]1,[62]2. WDR45, also known as WIPI4, is known to play important roles together with WDR45B in the process of autophagy, a critical cellular degradation and recycling pathway^[63]5–[64]8. Conventionally, BPAN pathogenesis has been attributed to autophagy dysfunctional caused by WDR45 mutations^[65]5,[66]6. However, emerging evidences suggest that WDR45 may also play critical roles beyond autophagy. Recent studies have revealed a more complex picture of BPAN pathogenesis. For instance, certain BPAN-related missense mutations produce neurodegenerative symptoms despite exhibiting near-normal autophagic activity, implying non-autophagic WDR45 functions are critical^[67]9. Furthermore, WDR45 has been implicated in preventing ferroptosis, a process distinct from autophagy, offering alternative explanations for BPAN pathogenesis^[68]10,[69]11. Additionally, WDR45 deficiency has been shown to disrupt ER homeostasis and phospholipid metabolism^[70]12,[71]13. These findings suggest that the neurodegenerative mechanisms underlying BPAN might involve non-autophagic functions of WDR45, highlighting the need for further investigation into its diverse cellular roles. Stress granules (SGs), dynamic cytoplasmic condensates formed via liquid-liquid phase separation (LLPS), are essential for regulating mRNA translation and stability during cellular stress^[72]14–[73]16. However, persistent SGs, which can form due to chronic stress or disease-related mutations, are linked to the pathogenesis of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), inclusion body myopathy (IBM), and Charcot-Marie-Tooth disease (CMT)^[74]17–[75]28. While short-lived SGs are rapidly disassembled, persistent SGs, are typically removed through autophagy-dependent degradation^[76]23,[77]29,[78]30. Despite their importance, the precise mechanisms governing SG disassembly remain poorly understood, hindering the development of targeted therapies for SG-related neurodegenerative diseases. Caprin-1 (Cell Cycle Associated Protein 1), a core RNA-binding protein in SGs, regulates mRNA stability and translation^[79]31. In addition to its involvement in SG biology, Caprin-1 is essential for neurodevelopment^[80]32–[81]34. While it promotes SG assembly through interactions with Ras-GTPase-activating protein-binding protein (G3BP), it is not essential for SG formation^[82]15,[83]35,[84]36. However, the precise role of Caprin-1 in regulating SG dynamics remains unclear. Given WDR45’s potential involvement in processes beyond autophagy and the critical role of SG in neurodegeneration, we investigated the interplay between WDR45 and SG dynamics. Here we demonstrate that WDR45 is recruited to SGs in response to various stress stimuli and is essential for SG disassembly through its interaction with Caprin-1. We show that WDR45 undergoes LLPS with Caprin-1 and possesses intrinsic gel-like properties that are crucial for SG disassembly. Furthermore, BPAN-associated mutations in WDR45 disrupt condensates formation and impair interaction with Caprin-1, leading to defective SG disassembly and pathological SGs accumulation. These findings provide mechanistic insights into BPAN pathogenesis, highlighting SG dysregulation as a potential therapeutic target in neurodegenerative diseases. Our study not only expands the understanding of WDR45 function but also reveals a layer of complexity in SG regulation, with broad implications for neurodegenerative disease research. Results WDR45 is incorporated into SGs and regulates SGs disassembly across different cell lines and under various stress conditions The dynamics of SGs are known to be disrupted in various neurodegenerative diseases. To explore the potential involvement of SG in BPAN, we examined whether WDR45 is recruited into SGs under stress conditions. Immunostaining with antibodies against endogenous WDR45 revealed a significant accumulation of WDR45 in cytoplasmic granules upon exposure to oxidative stress, heat shock (HS), or osmotic stress (Fig. [85]1a). These granules co-localized with established SGs markers G3BP1 in U2OS cells (Fig. [86]1a). Similarly, WDR45 was incorporated into SGs in human iPSC-induced neurons upon exposure to oxidative stress or heat shock (Fig. [87]1b and Supplementary Fig. [88]1a) and in SH-SY5Y upon oxidative stress (Supplementary Fig. [89]1b). These findings suggest that WDR45 recruitment into SGs is likely neither stress-specific nor cell type-dependent. Fig. 1. WDR45 regulates SG disassembly under various stress conditions. [90]Fig. 1 [91]Open in a new tab a Confocal images of U2OS cells treated with different stressors and stained with anti-G3BP1 (Red) and anti-WDR45 (Green) antibodies and DAPI (Blue) are shown. Stressors include sodium arsenite (SA, 500 μM, 1 h), heat shock (HS, 44 °C, 2 h), and osmotic stress (Sorbitol, 400 mM, 2 h). White boxes indicate the enlarged ROI and are displayed in separate channels (Zoom). Line graphs in the right panels show WDR45 and G3BP1 signals along the indicated white lines in the insets. Scale bar, 10 μm. b Representative confocal microscopy images of neural progenitor cells induced from induced pluripotent stem cells stained with anti-PAX6 (Red) (Left). Following the differentiation and maturation process, neural progenitor cells were induced into midbrain neurons which were stained with anti-TUJ1 (Red) (Right). Scale bar, 10 μm, and representative confocal images of G3BP1 (Red) and WDR45 (Green) in midbrain neurons treated with SA (SA, 500 μΜ, 45 min) and heat shock (HS, 1 h). Scale bar, 10 μm. The inset indicates the magnifying region of the white box. Inset scale bar, 1 μm. c WT, WDR45, WDR45B knockout cell lines and WDR45&WDR45B double knockout cell lines were untreated, treated with SA for 1 h, or recovered (Re) for 1.5 h, 2 h after SA removal. Confocal images of U2OS cells stained with anti-G3BP1 (Red) and DAPI (Blue) are shown. Scale bar, 10 μm. d Quantification of Cell with SGs percent (n = 3 independent experiments, every treatment cell number>200) under SA and at the recovery stage from stress as represented in (c). Data are shown as mean ± SD. ns, not significant, ****p < 0.0001 (two-way ANOVA with Tukey’s test). e WT, WDR45, WDR45B knockout cell lines and WDR45 WDR45B double knockout cell lines were untreated, treated with heat shock for 1 h, or recovered for 1.5 h, 2 h after removal of heat shock. Confocal images of U2OS cells treated with SGs induction stressors and stained with anti-G3BP1 (Red) and DAPI (Blue) are shown. Scale bar, 10 μm. f Quantification of Cell with SGs percent (n = 3 independent experiments, every treatment cell number>200) under heat shock and at the recovery stage from stress as represented in (e). Data were pooled from three independent experiments. Data are shown as mean ± SD. ns, not significant, ****p < 0.0001 (two-way ANOVA with Tukey’s test). Source data are provided as a Source Data file. Prompted by these findings, we next examined whether WDR45 regulates the dynamics of SGs. By silencing WDR45 using siRNA or employing CRISPR-mediated genetic knockout (KO) methods, we analyzed the dynamics of SGs induced by sodium arsenite (SA) or heat shock (HS) (Fig. [92]1c–f, Supplementary Figs. [93]1c–f and [94]2a–c). WDR45 KO in U2OS was validated through sequencing and immunostaining (Supplementary Fig. [95]1c, d). Intriguingly, genetic interference of WDR45 led to a significant delay in SG disassembly, while SG formation remained largely unaffected in the absence of WDR45 both across different cell lines, including Neuro-2a (N2a) and U2OS cells, and under alternative stress conditions, such as oxidative stress and heat shock (Fig. [96]1c–f, Supplementary Figs. [97]1e, f and [98]2a–c). Live-cell imaging of HeLa cells with G3BP1-GFP knock in under arsenite stress following WDR45 knock down also revealed the delayed SGs disassembly and fewer SGs per cell, though the size of the SGs remained unchanged (Supplementary Fig. [99]1g–j, Supplementary Movies [100]1–[101]4). To gain deeper insight into the role of WDR45 in SGs dynamics, we overexpressed WDR45 in U2OS cells. The results showed that overexpression of WDR45 did not influence SGs assembly upon arsenite stress (Supplementary Fig. [102]1k). We next investigated whether the other WIPI family proteins, namely WIPI1, WIPI2, and WDR45B play a similar role in SG disassembly as WDR45. We first examined their colocalization with SGs. GFP-tagged WDR45 but not WIPI1, WIPI2, or WDR45B colocalized with SGs (Supplementary Fig. [103]2d). Next, we performed siRNA-mediated silencing of WIPI1, WIPI2, WDR45B or ATG2A/2B. Among these, fonly the knockdown of WDR45B, a paralogue of WDR45, affected SG disassembly (Supplementary Fig. [104]2a–c). To further evaluate the role of WDR45B, we used CRISPR-mediated KO of WDR45B to deplete WDR45B or both WDR45 and WDR45B and evaluated the impact on SG dynamics in U2OS and N2a cells (Fig. [105]1c–f, Supplementary Fig. [106]1e, f and Supplementary Fig. [107]2a–c). While knockdown or KO of either WDR45 or WDR45B individually resulted in a substantial delay in SG disassembly, simultaneous silencing of both genes did not decelerate the delay (Fig. [108]1c–f, Supplementary Figs. [109]1e, f and [110]2a–c). Importantly, these effects were consistent across different cell lines and stress conditions (Fig. [111]1c–f, Supplementary Figs. [112]1e, f and [113]2a–c). Together, these results indicate that WDR45 and WDR45B have distinct, non-redundant roles in SG disassembly. Consistent with previous study by Ji et al. (2021)^[114]5, WDR45 and WDR45B single KO cells showed no significant autophagy defects (Supplementary Fig. [115]2e–g). These observations indicate that WDR45 regulates SG disassembly likely through autophagy-independent pathway. This finding aligns with prior studies that SGs undergo disassembly process independently of autophagy mechanisms^[116]29. We also supplemented the experiment of SGs dynamics under heat shock, while cells were pretreated with 20 µM bafilomycin (an autophagy inhibitor) for 18 h. Results demonstrated that bafilomycin treatment had no influence on SGs dynamics under this condition (Supplementary Fig. [117]2h, i). Given that WDR45 depletion may promote ER stress^[118]12, we confirmed that CRISPR-mediated knockout of WDR45 did not significantly alter the expression of HSPA5, indicating that WDR45 depletion does not permanently affect ER stress levels (Supplementary Fig. [119]2j, k). In contrast, siRNA-mediated knockdown of WDR45 transiently increased HSPA5 expression, potentially leading to ER stress (Supplementary Fig. [120]2l). Notably, defects in SG disassembly were observed in both WDR45 KO and KD cells (Fig. [121]1c–f, Supplementary Fig. [122]2b, c). Together, our results suggest that WDR45-dependent SG disassembly is mediated by distinct mechanisms separate from its autophagy-related or ER stress-related functions, highlighting a role for WDR45 in cellular stress responses. Caprin-1 forms condensates with WDR45 and facilitates its recruitment into SGs To gain insight into the mechanisms underlying the incorporation of WDR45 into SGs and its role in modulating disassembly, we analyzed the interactomes of WDR45 proteins in human cells under both physiological and stress conditions. WDR45-GFP was expressed in human U2OS cells, immunoprecipitated with anti-GFP nanobody, and co-purified proteins were analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS) (Fig. [123]2a, Supplementary Table [124]1). Fig. 2. WDR45 interacts with Caprin-1 and undergoes LLPS with Caprin-1. [125]Fig. 2 [126]Open in a new tab a Comparative proteomic analysis of GFP (control group) and WDR45-GFP (experimental group) under SA treatment and untreated conditions. Cells expressing either GFP or WDR45-GFP were treated with SA for 1 hour or left untreated as controls. Each experimental condition was performed in triplicate to ensure reproducibility and statistical robustness (Created with Biorender.com). b Venn diagrams comparing the proteomes of CON-WDR45 and SA-WDR45 with the core components of SGs. The green box highlights the shared proteins between CON-WDR45 proteome and the SG core components. The orange box highlights the shared proteins between SA-WDR45 proteome and the SG core components. c Western blot analysis of immunoprecipitation (IP) experiments using GFP (control), WDR45-GFP, and WDR45B-GFP. IP was performed using antibodies against GFP to assess the interaction with Caprin-1. d Representative confocal images of U2OS cells under control and SA treatment (500 μM 45 min) stained with anti-Caprin-1 (Red) and anti-WDR45 (Green). DAPI is shown in blue. Scale bar, 10 μm. e Representative confocal images of U2OS WT and CAPRIN1 KO cells under SA treatment (500 μM 45 min) stained with anti-G3BP1 (Red) and anti-WDR45 (Green). DAPI is shown in blue. Scale bar, 10 μm, Zoom’s Scale bar, 1 μm. f Representative SIM (Structure Illumination Microscopy) images of G3BP1-positive SGs (Red) and WDR45 (Green) in WT and CAPRIN1 KO U2OS cells. Scale bar, 1 μm, Zoom’s Scale bar, 0.5 μm. g Scatterplot representing the co-localization correlation between G3BP1 and WDR45 signal (WT and CAPRIN1 KO cells) during the SA stage as represented in (f). Data are shown as mean ± SD. Statistical significance was determined using an unpaired and two-sided t-test comparison. Each cell represents the Pearson coefficient analysis of cell ROI. ****P < 0.0001. n = 20 from 3 independent experiments. h Relative fluorescence intensity of WDR45-GFP before and after photobleaching over time. Data are represented as mean ± SEM for n = 7. i Dynamics of WDR45 foci indicated by the yellow circle in U2OS cells stably transfected with WDR45-GFP were analyzed using FRAP. Representative images of the same foci before and after photobleaching are shown. Scale bar, 1 μm. j Phase diagram of WDR45 in vitro. k 1,6-hexanediol (1,6-HD) destroys the WDR45 condensates in vitro. Scale bar, 10 μm. l LLPS of purified recombinant G3BP1 (Cyan), Caprin-1 (Red), and WDR45 (Green) in vitro. Recombinant proteins were mixed pairwise at a 1:1 molar ratio (Caprin-1 + WDR45, G3BP1 + Caprin-1, G3BP1 + WDR45) to observe LLPS under physiological conditions with 150 mM NaCl. Scale bar, 10 μm. m Dynamics of WDR45 (or +Caprin-1) foci (Green) indicated by yellow circle in vitro were analyzed using FRAP. Representative images of the same foci before and after photobleaching are shown. Scale bar, 5 μm. n Relative fluorescence intensity of WDR45 (or +Caprin-1) before and after photobleaching over time. Data are represented as mean ± SEM. ****p < 0.0001 by paired and two-sided t-test. n = 17 for WDR45+Caprin-1, n = 7 for WDR45 alone. Source data are provided as a Source Data file. Our results indicated significant changes in WDR45-interacting proteins under stress conditions. While 300 interactors were identified under normal conditions, this number was reduced to 79 following arsenite treatment (Fig. [127]2b). This suggests that WDR45 is responsive to cellular stress. Subsequently, we performed pathway enrichment analysis using the online tool ShinyGo^[128]37. The analysis revealed that pathways associated with neurodegenerative diseases—such as Amyotrophic Lateral Sclerosis, Alzheimer’s disease, Huntington’s disease, and Parkinson’s disease—were enriched across both conditions (Supplementary Fig. [129]3a). These findings underscore the potential role of WDR45 in neurodegenerative diseases. Comparing the WDR45 interactome under normal conditions and stress conditions with the core SGs network published by Yang^[130]15, we identified Caprin-1 as the only overlapping protein (Fig. [131]2b). We confirmed the interaction between WDR45 and Caprin-1 using co-immunoprecipitation and Glutathione S-transferase (GST) pull-down (Fig. [132]2c and Supplementary Fig. [133]7c). Furthermore, WDR45 co-localized with Caprin-1 under oxidative stress in U2OS cells, iPSC-derived neurons and SH-SY5Y (Fig. [134]2d and Supplementary Fig. [135]3b, c). In addition, WDR45B also interacted with Caprin-1 (Fig. [136]2c). To study the WDR45 interactors more comprehensively, we also confirmed the interaction between WDR45 and NUP98 and G3BP1, with no detectable binding observed under standard assay conditions. (Supplementary Fig. [137]3d). Given the critical role of Caprin-1 in the core network of stress granules, Caprin-1 was selected for subsequent analysis. To determine whether WDR45 incorporation into SGs is dependent on Caprin-1, we generated a CAPRIN1 KO cell line using the CRISPR-Cas9, which showed complete loss of Caprin-1 expression (Supplementary Fig. [138]3e). The incorporation of WDR45 into SGs was significantly reduced in CAPRIN1 KO cells (Fig. [139]2e). The result demonstrated that Caprin-1 is essential for WDR45 recruitment to SGs. Super-resolution structured illumination microscopy (SIM) further confirmed this finding. In WT cells, WDR45 showed a high degree of co-localization with SGs, as indicated by the high Pearson correlation coefficient (Fig. [140]2f, g). However, in CAPRIN1 KO cells, the Pearson correlation coefficient between WDR45 and SGs was significantly reduced, suggesting that the recruitment of WDR45 to SGs is impaired in the absence of Caprin-1 (Fig. [141]2f, g). The enrichment of WDR45 in SGs promoted us to investigate the dynamics of WDR45 puncta in the cytosol using fluorescence recovery after photobleaching (FRAP). WDR45-GFP puncta in cytosol were highly dynamic, recovering and plateauing within 60 s post-photobleaching (Fig. [142]2h, i). Caprin-1 has been shown to undergo LLPS with RNA, G3BP1, or FMRP, but not individually^[143]14,[144]33,[145]36,[146]38. We hypothesized that WDR45 might form droplets in vitro or modulate the LLPS properties of Caprin-1. To test this, we purified MBP-HIS-WDR45 (hereafter WDR45) and MBP-HIS-mCherry-Caprin-1 (hereafter Caprin-1) and removed the MBP-HIS tag using 3 C protease for in vitro phase separation assay. The cellular concentration of WDR45 is 34 nM^[147]39. At physiological concentrations, WDR45 formed condensates in vitro without requiring any crowding agent (Fig. [148]2j, Supplementary Fig. [149]3f). This observation is consistent with WDR45 being one of the top-scored phase separation candidates according to the DosPS predictor^[150]40, despite its relatively low percentage of disordered regions. However, in the presence of RNA or PEG, WDR45 protein produced amorphous, fiber-like aggregates (Supplementary Fig. [151]3g). We further employed 1,6-hexanediol (1,6-HD), which inhibits phase separation by disrupting multivalent hydrophobic protein-protein interactions^[152]17. These WDR45 condensates were no longer observed in the presence of 1,6-HD, indicating that the hydrophobic interactions drive WDR45 condensates formation (Fig. [153]2k). Consistent with previous reports, Caprin-1 did not form droplets individually but did undergo LLPS with G3BP1, RNA, or in the presence of 10% PEG (Fig. [154]2l and Supplementary Fig. [155]3g). When mixed with purified full-length WDR45, Caprin-1 formed condensates with WDR45, while WDR45 did not form condensates with G3BP1 (Fig. [156]2l and Supplementary Fig. [157]4a). With increasing WDR45 concentration, WDR45/Caprin-1 condensate correspondently grew in size, demonstrating the concentration-dependent nature of WDR45/Caprin-1 condensate (Supplementary Fig. [158]4b, c). Notably, purified WDR45B did not form condensate, even with Caprin-1, which is consistent with the observation that WDR45B did not form puncta under stress conditions (Supplementary Fig. [159]2d, [160]4a). Next, using FRAP on in vitro WDR45 condensates with or without Caprin-1, we found that the FRAP ratio of WDR45 condensates without Caprin-1 was approximately 10%, significantly lower than that with Caprin-1 (∼60%) (Fig. [161]2m, n). This difference in FRAP ratios indicates that WDR45 homotypic interaction-mediated gel-like phase separation, while heterotypic interactions with Caprin-1 promote a transition to a more dynamic, liquid-like state. Consistently, WDR45/Caprin-1 droplet fused completely within 12 seconds, whereas WDR45 alone exhibited slower fusion rates (Supplementary Fig. [162]4d, e). These results consistent with the observation that WDR45 interacted with Caprin-1 and formed dynamic puncta in cell culture (Fig. [163]2c, d, h, i). These findings demonstrate that Caprin-1 plays a critical role in modulating the dynamic properties of WDR45 condensates, facilitating a transition from a static, gel-like state to a dynamic, liquid-like phase. This transition likely enables WDR45 to rapidly respond to cellular stress, highlighting its functional versatility in SG dynamics and stress responses. WDR45 competes with G3BP1 for Caprin-1 binding to facilitate SG disassembly To investigate whether the interaction between WDR45 and Caprin-1 is critical for SG disassembly, we examined their binding dynamics during stress and recovery. Cells stably expressing WDR45-GFP were subjected to stress and monitored during recovery at 1.5 and 2 hours. The interaction between WDR45-GFP and Caprin-1 increased significantly upon stress, peaking at 1.5 hours of recovery (Fig. [164]3a, b). This suggests that WDR45 binds more tightly to Caprin-1 during stress and recovery phases, potentially facilitating SG disassembly. Fig. 3. WDR45 competes with G3BP1 for Caprin-1 binding to facilitate SG disassembly. [165]Fig. 3 [166]Open in a new tab a Western blot analysis of immunoprecipitation (IP) experiments examining the Caprin-1 enrichment capability in U2OS cells expressing WDR45-GFP under different conditions: control (untreated), SA treatment for 1 hour (SA 1 h), and recovery periods of 1.5 hours and 2 hours post-treatment. (n = 3 independent experiments). b Quantification of Caprin-1 enrichment ability with WDR45-GFP under SA treatment and during recovery stages, as represented in (a). Data were pooled from three independent experiments and normalized to the control condition. Results are presented as mean ± SD, with statistical significance indicated by *p < 0.05 and **p < 0.01, determined by one-way ANOVA with Tukey’s test. c WT, WDR45, CAPRIN1 knockout cell lines and WDR45&CAPRIN1 double knockout cell lines were untreated, treated with SA for 1 h, or recovered for 1.5 h, 2 h after removal of SA. Confocal images of U2OS cells treated with SGs induction stressors and stained with anti-G3BP1 (Green) and DAPI (Blue) are shown. Scale bar, 10 μm. d Quantification of Cell with SGs percent under SA (n = 3 independent experiments) and at the recovery stage from stress as represented in (c). Data were pooled from three independent experiments. Data are shown as mean ± SD. ns, not significant, ****p < 0.0001 (two-way ANOVA with Tukey’s test). e Western blot analysis of endogenous immunoprecipitation (IP) performed with G3BP1 to assess the interaction with Caprin-1 in wild-type (WT) and WDR45 knockout (KO) cells. The enrichment ability of G3BP1 for Caprin-1 was assessed under two conditions: control (untreated) and SA treatment (SA). (n = 3 independent experiments). f Quantification of G3BP1’s ability to enrich Caprin-1 under control and SA treatment conditions, as shown in (e). Data were pooled from three independent experiments and normalized to Caprin-1 enrichment in WT cells under the control condition. Results are presented as mean ± SD, with statistical significance indicated by *p < 0.05, determined by one-way ANOVA with Tukey’s test. g Scatterplot representing the co-localization correlation between G3BP1 and Caprin-1 signal (WT and WDR45 KO cells) during the SA stage. Data are shown as mean ± SD. Statistical significance was determined using an unpaired and two-sided t-test. ***p < 0.001. n = 20 from 3 independent experiments. h Representative SIM (Structure Illumination Microscopy) images of G3BP1-positive SGs (Red) and Caprin-1 (Green) in WT and WDR45 KO U2OS cells. Scale bar, 1 μm, Zoom’s Scale bar, 0.5 μm. (i), LLPS of purified recombinant G3BP1 (Cyan), Caprin-1 (Red), and WDR45 (Green) in vitro. Keep the concentrations of G3BP1 and Caprin-1 constant while gradually increasing the concentration of WDR45 to observe LLPS under physiological conditions with 150 mM NaCl. LLPS was visualized using confocal microscopy. Scale bar, 10 μm. j LLPS of purified recombinant G3BP1 (Cyan), Caprin-1 (Red), and WDR45 (Green) in vitro. Keep the concentrations of WDR45 and Caprin-1 constant while gradually increasing the concentration of G3BP1 to observe LLPS under physiological conditions with 150 mM NaCl. Scale bar, 10 μm. Source data are provided as a Source Data file. To explore whether WDR45 modulates SG dissolution through a Caprin-1-dependent pathway, we generated CAPRIN1 single KO cells and WDR45&CAPRIN1 dKO cells, then monitored the dynamics of SGs induced by SA or HS. CAPRIN1 KO rescued the dissolution defect observed in WDR45 KO cells (Fig. [167]3c, d and Supplementary Fig. [168]5a, b). Interestingly, genetic interference with CAPRIN1 individually led to faster dissolution of SGs, whereas SGs formation was largely unaffected in the absence of Caprin-1 under both stress conditions (Fig. [169]3c, d, Supplementary Fig. [170]5a, b). We also conducted the FRAP assay in WT and WDR45 KO cells to exam whether WDR45 regulates SGs mobility. The mobility of GFP-G3BP1 puncta remained unchanged following WDR45 knockout, suggesting that WDR45 does not regulate SG dissolution by altering SG mobility (Supplementary Fig. [171]5f, g). In addition, WDR45 depletion did not affect the protein levels of G3BP1 or Caprin-1 (Supplementary Fig. [172]5c–e). These findings indicate that WDR45 modulates SG dissolution through a Caprin-1-dependent pathway. SGs assemble through a core protein-RNA interaction network, with G3BP serving as the central node of this network^[173]15. Based on our observations, we hypothesized that WDR45 competes with G3BP1 for Caprin-1 binding, thereby modulating the SG core network and dynamics. To test this hypothesis, we first validated that the interaction between G3BP1 and Caprin-1 increased during stress and decreased during recovery, using a HeLa G3BP1-GFP knock-in cell line (Supplementary Fig. [174]5h). In contrast, the interaction between WDR45-GFP and Caprin-1 increased during recovery, peaking at 1.5 h post-stress (Fig. [175]3a, b). To further investigate this competition, we performed Co-IP in WDR45 KO cells and G3BP1/2 dKO cells. Interestingly, the interaction between G3BP1 and Caprin-1 significantly enhanced in WDR45 KO cells under both control and stress conditions (Fig. [176]3e, f). Conversely, the interaction between WDR45 and Caprin-1 significantly enhanced in G3BP1/2 dKO cells under both conditions (Supplementary Fig. [177]5i). Given that previous studies have shown that the interaction between G3BP1 and Caprin-1 is RNA-dependent^[178]36, we next examined the interaction between WDR45 and Caprin-1 following RNase treatment. Following RNase treatment, we observed a significant enhancement in the interaction between WDR45 and Caprin-1 (Supplementary Fig. [179]5j). Furthermore, we mapped the interaction domain of WDR45 and Caprin-1 to the NTD-GIM domain of Caprin-1, which is also responsible for its interaction with G3BP1^[180]36 (Supplementary Fig. [181]5k, l). Together, these findings demonstrate that WDR45 competes with G3BP1 for binding to Caprin-1, providing a mechanistic basis for its role in modulating SG dynamics. To further confirm this finding, we conducted SIM-based super-resolution imaging. WDR45 KO cells exhibited an increased Pearson correlation coefficient between G3BP1 and Caprin-1 (Fig. [182]3g, h). Moreover, SIM-based super-resolution imaging revealed that the molecular density of G3BP1 in SGs significantly increased in WDR45 KO cells, whereas it was reduced in CAPRIN1 KO cells (Supplementary Fig. [183]5m, n). These results also suggest that WDR45 and Caprin-1 have opposing roles in regulating the SG core, with WDR45 KO leading to a strengthened and more rigid SG core, while CAPRIN1 KO results in a looser and less stable core. We next asked whether WDR45 and G3BP1 compete directly for Caprin-1 binding in vitro. In GST pull-down assays, purified GST–Caprin-1 efficiently bound WDR45, but the addition of purified G3BP1 markedly reduced this interaction (Supplementary Fig. [184]5o). To further determine whether the competition is dose-dependent, we conducted in vitro phase separation experiments using three purified proteins. WDR45 could undergo phase separation with G3BP1 and Caprin-1 together to form droplets. As the concentration of WDR45 increased, G3BP1 was excluded from the droplets (Fig. [185]3i). Conversely, increasing the concentration of G3BP1 excluded WDR45 from the droplets (Fig. [186]3j). Notably, high concentration of WDR45 led to irregularly shaped droplets, indicating potentially gel-like condensates (Fig. [187]3i). FRAP assay showed that increasing concentrations of WDR45 reduced droplet mobility, consistent with the formation of gel-like condensates (Supplementary Fig. [188]5p, q). Previous studies have shown that stress granules contain a stable core structure^[189]30 and SG assembly is driven by a core protein-RNA network^[190]15. Our findings reveal a critical regulatory mechanism underlying SG dynamics: WDR45 competes with G3BP1 for Caprin-1 binding, loosening the SG core and facilitating SG disassembly during the recovery process. This competition highlights the essential interplay between WDR45, Caprin-1, and G3BP1 in destabilizing the SG core network and regulating SG dynamics. The WD5 domain of WDR45 mediates condensates formation and SG disassembly WDR45, a member of the WIPI protein family, has a relatively simple structure composed of seven WD (tryptophan-aspartate) domains with significantly varying amino acid compositions (Fig. [191]4a). To identify the domains responsible for WDR45’s function in SGs, we systematically deleted segments of its WD domain (Fig. [192]4a and Supplementary Fig. [193]6a). Fig. 4. WD5 domain in WDR45 mediates LLPS with Caprin-1. [194]Fig. 4 [195]Open in a new tab a Schematic diagram of WDR45 domain structure and amino acid sequence of each domain. Truncations of ΔWD1-4, ΔWD5-7, ΔWD5-6, ΔWD6-7, ΔWD7, ΔWD6, ΔWD5 are shown. b Western blot analysis of immunoprecipitation (IP) experiments examining the Caprin-1 enrichment capability in U2OS cells expressing GFP, WDR45-GFP and its truncations under control (untreated). c LLPS of purified recombinant Caprin-1 (Red), WDR45 and its truncations (Green) in vitro. Recombinant proteins were mixed pairwise at 1:1 molar ratio to observe LLPS under physiological conditions with 150 mM NaCl. Scale bar, 10 μm. d Confocal images of U2OS cells (expressing GFP, WDR45-GFP and its truncations) that were treated with SA and stained with anti-G3BP1 (Red) antibody and DAPI (Blue) are shown. Scale bar, 10 μm. e Confocal images of WDR45 KO U2OS cells (expressing GFP, WDR45-GFP and its truncations) that were treated with SA and recovery 2 h stained with anti-G3BP1 (Red) antibody and DAPI (Blue) are shown (n = 3 independent experiments, every treatment cell number>200). Scale bar, 10 μm. f Quantification of Cell with SGs percent under SA, at the recovery stage from stress as represented in (e). Data were pooled from three independent experiments. Data are shown as mean ± SD. ns, not significant, **p < 0.01, ****p < 0.0001 (one-way ANOVA with Tukey’s test). Source data are provided as a Source Data file. Initial analysis of purified truncated proteins revealed that the ΔWD1-4 segment retained condensation properties, whereas ΔWD5-7 did not, indicating that the WD5-7 region is critical for WDR45 phase separation (Supplementary Fig. [196]6a). Further truncation experiments with ΔWD7, ΔWD6-7, ΔWD5-6, ΔWD5, and ΔWD6 variants demonstrated that the WD5 and WD6 domains are essential for mediating WDR45 phase separation (Supplementary Fig. [197]6a). Specifically, deletion of WD6 partially inhibited phase separation, whereas deletion of WD5 completely abolished it (Supplementary Fig. [198]6a). Notably, the isolated WD5 domain formed aggregates in vitro, suggesting that WD5 mediates the gel-like properties of WDR45, while other domains may stabilize or enhance WD5’s phase separation capability (Supplementary Fig. [199]6b). To further investigate the role of WD5 in WDR45’s interaction with Caprin-1, we performed Co-IP assays. Truncated variants lacking WD5 (ΔWD5 and ΔWD5-6) lost the ability to interact with Caprin-1, whereas variants lacking only WD6 (ΔWD6) retained this interaction both in cell culture and in vitro (Fig. [200]4b, Supplementary Fig. [201]6c). Consistently, ΔWD5 variants failed to form droplets with Caprin-1 in vitro, highlighting the importance of WDR45 gel-like condensates in facilitating Caprin-1 LLPS (Fig. [202]4c). Given that WDR45 forms condensates with Caprin-1 via the WD5 domain, we next examined whether WDR45’s incorporation into SGs also depends on WD5. Using U2OS cell lines expressing WDR45-GFP with deletions in WD1-4 (ΔWD1-4) or WD5-7 (ΔWD5-7), we observed that under stress conditions, WDR45-GFP(ΔWD5-7) failed to localize to SGs, whereas WDR45-GFP(ΔWD1-4), which retains phase separation properties, successfully incorporated into SGs (Fig. [203]4d). Further analysis of ΔWD5 and ΔWD6 variants confirmed that deletion of WD5, but not WD6, abolished SG localization (Fig. [204]4d). To assess the functional relevance of WD5 in SG disassembly dynamics, we examined WDR45 KO cells. WDR45-GFP(ΔWD5) failed to rescue SG disassembly, whereas both WDR45-GFP(ΔWD6) and wild-type WDR45 successfully restored normal SG dynamics (Fig. [205]4e, f). To further explore the role of WD5, we analyzed a truncated version of WDR45 (Δ171–191, termed “del peak”) predicted using the FuzDrop online tool ([206]https://fuzdrop.bio.unipd.it/predictor)^[207]41 (Supplementary Fig. [208]6d). The del peak protein was unable to form condensates, suggesting that this region is critical for phase separation (Supplementary Fig. [209]6e). Previous studies have shown that substituting serine (S) with proline (P) can significantly alter the functional properties of various protein domains, particularly in highly conserved serine residues within WD repeat domains, leading to structural changes^[210]42. Based on this, we focused on serine 183 (S183), located within the overlapping region of the WD5 domain and the peak. By generating S183P and S183A mutants, we discovered that mutations at S183 abolished WDR45’s ability to form condensates, weakened its interaction with Caprin-1, and failed to rescue SGs disassembly defects in WDR45 KO cells (Supplementary Fig. [211]6e–h). These findings demonstrate that the WD5 domain, particularly S183, is critical for condensate formation, mediating the interaction between WDR45 and Caprin-1, and regulating SG disassembly. This highlights the importance of the WD5 region in maintaining proper SG dynamics and provides insights into the molecular mechanisms underlying WDR45 function. BPAN-associated mutations in WDR45 impair phase separation and SG disassembly Seven female patients developed neurodegeneration symptoms and there was no clear correlation between the autophagic activity and the age of symptoms onset^[212]9. To assess whether BPAN-associated mutations in WDR45 directly affect its phase separation properties, we constructed and purified proteins for following eight BPAN-related WDR45 mutations associated with neurodegeneration symptoms at known ages: R13P, N61K, L98P, N202K, G204D, A209D, S210P and S251del (Fig. [213]5a). These mutant proteins exhibited a marked sensitivity to both salt and protein concentration for condensate formation (Supplementary Fig. [214]7a, b). Compared to wild-type WDR45, all BPAN-related mutations significantly reduced condensate formation capacity, with mutations in the WD5 domain (N202K, G204D, A209D, and S210P) reducing condensate formation capacity to only 10–20% of wild-type levels. (Fig. [215]5a, b, Supplementary Fig. [216]7a, b). Fig. 5. BPAN-associated mutations in WDR45 impair its function in SG disassembly. [217]Fig. 5 [218]Open in a new tab a Schematic diagram of β-propeller protein-associated neurodegeneration (BPAN) related WDR45 mutations’ domain structure. b The phase separation ability of respective WDR45 mutants was graphed alongside the age of onset for neurodegenerative symptoms. The age of onset for these symptoms is provided in parentheses after each mutation. The Pearson’s correlation coefficient (R) reflecting the correlation between the extent of condensates formation ability defects and the age of onset for these symptoms is presented. c Western blot analysis of Co-IP experiments using GFP-trap to assess the interaction between WDR45-GFP, and its mutations with Caprin-1. d LLPS of purified recombinant Caprin-1 (Red), WDR45 and its mutations (Green) in vitro. Recombinant proteins were mixed pairwise at 1:1 molar ratio to observe LLPS under physiological conditions with 150 mM NaCl. Scale bar, 10 μm. e Quantification of droplets area for Caprin-1 and WDR45 and its mutations as represented in (d). Data are shown as mean ± SD. ns, not significant, ****p < 0.0001 (one-way ANOVA with Tukey’s test). WT: n = 72; R13P: n = 63; L98P: n = 43; N202K: n = 45; G204D: n = 49; A209D: n = 51; S210P: n = 52; S251 del: n = 56 from 3 independent experiments. f Confocal images of WT U2OS cells (expressing GFP, WDR45-GFP and its mutations) that were treated with SA and stained with anti-G3BP1 (Red) antibody and DAPI (Blue) are shown. Scale bar, 10 μm. g Summary of WDR45 and its mutants in relation to SG localization. h Confocal images of WDR45 KO U2OS cells (expressing GFP, WDR45-GFP and its mutations) that were treated with SA and recovery 2 h stained with anti-G3BP1 (Red) antibody and DAPI (Blue) are shown (n = 3 independent experiments, every treatment cell number>200). Arrowheads indicate the successfully transfected cells. Scale bar, 10 μm. i Quantification of Cell with SGs percent under SA, at the recovery stage from stress as represented in (h). Data were pooled from three independent experiments. Data are shown as mean ± SD. ns, not significant, ***p < 0.001, ****p < 0.0001 (one-way ANOVA with Tukey’s test). Source data are provided as a Source Data file. These findings directly demonstrate that BPAN-associated WDR45 mutations impair the protein’s ability to form condensate, likely disrupting its physiological role. Given that BPAN is characterized by severe neurodegeneration, we explored whether the degree of condensation impairment correlates with disease progression. Linear regression analysis revealed a strong positive correlation between the age of BPAN onset and the extent of phase separation impairment (Pearson correlation coefficient R = 0.598) (Fig. [219]5b). Mutations associated with earlier disease onset exhibited the most severe defects in phase separation capacity (Fig. [220]5b). These results suggest that WDR45’s condensate formation capability is closely tied to BPAN progression, potentially through interference with its role in SG dynamics. Further analysis confirmed that these mutant WDR45 proteins exhibited reduced condensate formation properties and failed to interact with Caprin-1 both in cell culture and in vitro (Fig. [221]5c, Supplementary Fig. [222]7c). Additionally, these mutations showed significantly reduced ability to promote Caprin-1 phase separation (Fig. [223]5d, e). To investigate whether BPAN-associated WDR45 mutations affect SG dynamics, we introduced these mutations into WDR45-GFP constructs. Under stress conditions, mutations within the WD5 domain (N202K, G204D, A209D, S210P, and S251del) failed to localize to SGs and were unable to rescue disassembly defects in WDR45 KO cells (Fig. [224]5f–i, Supplementary Fig. [225]7d). Notably, these mutations even exacerbated the disassembly defects (Fig. [226]5h, i). In contrast, mutations R13P and L98P, which retained phase separation properties, successfully incorporated into SGs and rescued SG disassembly defects (Fig. [227]5f–i and Supplementary Fig. [228]7d). These findings directly link WDR45’s role in SG disassembly to BPAN pathology, underscoring the importance of its interaction with Caprin-1 in SG dynamics. The inability of BPAN-associated mutations to mediate phase separation and promote SG disassembly highlights a critical mechanism underlying disease progression. WDR45 deletion exacerbates accumulation of pathological SGs Pathological SGs, induced by disease-associated factors, are characterized by their impaired dynamics, reduced disassembly, and harmful effects on cellular function^[229]28. Factors driving the formation of these pathological SGs include mutant proteins such as FUS and dipeptide-repeat proteins (DPRs). To assess the broader implications of WDR45 in neurodegenerative diseases, we firstly examined its role in the accumulation of pathological SGs containing ALS-associated mutant FUS-R521C protein. The FUS-R521C mutant is known to translocate into SGs and contributes to disease pathology^[230]43–[231]45. Plasmids encoding either wild-type FUS (FUS WT) or the FUS-R521C were transfected into WT and WDR45 KO cells. Forty-eight hours post-transfection, we observed a significant increase of FUS-R521C-positive SGs in WDR45 KO cells compared to WT cells following transfection (Fig. [232]6a–d). Moreover, the FUS-R521C-positive SGs in WDR45 KO cells were approximately three times larger than those in WT cells, indicating a pronounced accumulation of pathological SGs in the absence of WDR45 (Fig. [233]6a–d). Fig. 6. The accumulation of ALS-related pathological SGs in WDR45 KO cells. [234]Fig. 6 [235]Open in a new tab a WT and WDR45 KO cells under normal conditions were transfected with plasmids encoding HA-tagged wild-type FUS (FUS WT) and FUS-R521C (FUS R521C) and stained for HA and TIAR (a SG marker). Scale bar, 10 μm. b Quantification of Cell with FUS-positive SGs percent under control as represented in (a). Data are shown as mean ± SD. ***p < 0.001 (unpaired and two-sided t-test) n = 3 independent experiments. c WT and WDR45 KO cells under normal conditions were transfected with plasmids encoding HA-tagged wild-type FUS (FUS WT) and FUS-R521C (FUS R521C) and stained for HA and TIAR. Scale bar, 10 μm. d Quantification of area for FUS-positive SGs. Scatter plot showing the cytoplasmic FUS area (μm^2) in WT (mean=0.705 μm^2, n = 147) and WDR45 KO (mean=2.797 μm^2, n = 143) cell lines. ****p < 0.0001 (unpaired and two-sided t-test) n = 3 independent experiment. e WT and WDR45 KO cells under normal condition were transfected with plasmid encoding GFP-tagged GR50 and stained for G3BP1. White arrowheads indicate the GR-positive SGs. Scale bar, 10 μm. Zoom’s scale bar, 10 μm. f Quantification of Cell with GR50-SGs percent under control as represented in (e). Data are shown as mean ± SD. ***p < 0.001 (unpaired and two-sided t-test) n = 3 independent experiment. Source data are provided as a Source Data file. Additionally, we examined the impact of poly(GR) repeats associated with the ALS/FTD gene C9orf72, which are known to disrupt SG dynamics^[236]18,[237]46. When GR50 was transfected into cells, U2OS WDR45 KO exhibited a higher percentage of GR-positive SGs compared to WT cells (Fig. [238]6e, f). These findings highlight WDR45’s protective role in regulating SG dynamics and mitigating the formation of pathological SGs associated with neurodegenerative diseases. The loss of WDR45 appears to exacerbate pathological protein aggregation, suggesting a potential link between WDR45 dysfunction and the progression of neurodegenerative disorders. iPSC-derived midbrain neurons from WDR45-mutant patient exhibited delayed SG recovery To establish a disease model for investigating the molecular mechanisms underlying WDR45-related disorders, we previously derived iPSCs from a female BPAN patient carrying a non-canonical splice site mutation in the WDR45 gene (c.827+1 G > A), which encodes a truncated version of WDR45 termed p.(Ala276AlafsTer1) ([239]https://hpscreg.eu/cell-line/CIPi005-A)^[240]47 (Supplementary Fig. [241]8a). No obvious defects in autophagy flux were detected in the patient-derived iPSCs, which indicates that this mutation may cause some defect in a non-autophagic function (Supplementary Fig. [242]8b, c). To explore the role of WDR45 in SG dynamics in neuronal model, the derived iPSCs were differentiated into neural progenitor cells and subsequently into midbrain neurons (Supplementary Figs. [243]1a, [244]8d, [245]e). The induced neurons displayed normal autophagy levels, as indicated by comparable SQSTM1 and LC3B expression between patient and control neurons (Supplementary Fig. [246]8f). Additionally, there was no significant difference in endoplasmic reticulum (ER) stress between the two groups, as shown by similar HSPA5 expression levels (Supplementary Fig. [247]8g). Interestingly, midbrain neurons derived from the patient exhibited significantly slower SG recovery following arsenite-induced stress compared to healthy control neurons, highlighting the critical role of WDR45 in SG disassembly (Fig. [248]7a, b). To further investigate the molecular basis of this defect, we purified the truncated p.(Ala276AlafsTer1) protein and found that it was unable to form condensates with Caprin-1 and also failed to associate with Caprin-1, as shown by Co-IP and GST pull-down assays. (Fig. [249]7c, Supplementary Fig. [250]8h, i). Furthermore, plasmids encoding the p.(Ala276AlafsTer1) could not colocalize with SGs (Supplementary Fig. [251]8j) and failed to rescue the SG disassembly defect in WDR45 KO cell line (Fig. [252]7d, e). This finding supports the notion that BPAN-associated mutations compromise WDR45’s function in SG dynamics by disrupting its phase separation property and interaction with Caprin-1. Fig. 7. iPSC-derived midbrain neurons from WDR45-mutant patient show delayed SG recovery. [253]Fig. 7 [254]Open in a new tab a Representative confocal images of neurons from patient and age-matched control. Neurons were subjected to SA stress for 30 min and recovered for 120 min and 150 min, and then stained with anti-Caprin-1 (Red) and anti-TUJ1 (Green). Nuclei were stained with DAPI. Scale bar, 10 μm. Zoom’s scale bar, 10 μm. b Statistical results of cells with SGs under different treatments as represented in (b). Data were performed with three biological repeats. Data are presented as mean ± SD. ns, not significant, **p < 0.01 by two-way ANOVA. c LLPS of purified recombinant Caprin-1 (Red), WDR45 Patient (Green) in vitro. Recombinant proteins were mixed pairwise at a 1:1 molar ratio to observe LLPS physiological conditions with 150 mM NaCl. Scale bar, 10 μm. d Confocal images of WDR45 KO U2OS cells (expressing GFP, p(Ala276AlafsTer1)) that treated with SA following recovered for 120 min, and stained with anti-G3BP1 (Red) antibody and DAPI (Blue) are shown, n = 3 independent experiment. Scale bar, 10 μm. e Quantification of Cell with SGs percent under SA, at the recovery stage from stress as represented in (d). Data were pooled from three independent experiments. Data are shown as mean ± SD. ns, not significant (unpaired and two-sided t-test). f A proposed model of WDR45-mediated SG disassembly. Functional WDR45 helps SGs disassembly through competing with G3BP1 for Caprin-1, thus weakening the SGs core network. While mutated WDR45 (WDR45 KO or BPAN-related WDR45 mutations) failed to interact with Caprin-1, and causing SGs disassembly defect (Created with Biorender.com). Source data are provided as a Source Data file. Discussion This study identifies a function for WDR45 in SG disassembly and advances our understanding of SG dynamics regulation and BPAN pathogenesis (Fig. [255]7f). We demonstrate that WDR45, via its WD5 domain, forms gel-like condensates and undergoes LLPS with Caprin-1 (Fig. [256]2l). This interaction is essential for efficient SG disassembly. Critically, BPAN-associated mutations, particularly those within WD5, disrupt WDR45’s phase separation capacity and its interaction with Caprin-1, leading to defective SG disassembly (Fig. [257]5d–i). This mechanistic defect correlates with earlier disease onset in BPAN patients, directly implicating impaired SG dynamics in disease pathogenesis (Fig. [258]5b). Furthermore, the distinct roles of WDR45’s WD5 (SG disassembly) and WD3 (ATG2B binding for autophagy) domains^[259]48 highlight the functional diversity of this protein and underscore the specificity of our findings. We emphasize that various disease-related mutations in WDR45 contribute to BPAN through divergent molecular mechanisms. Some mutations primarily affect autophagy, such as R13P and L98P^[260]5, while others compromise WDR45’s role in SG disassembly or selectively affect WDR45’s ability to undergo phase separation (Fig. [261]5h, i, Supplementary Fig. [262]7a, b). Our findings reveal that WDR45 competes with G3BP1 for Caprin-1 binding, thereby modulating the core SG network. This competition weakens the G3BP1-Caprin-1 interaction, which in turn promotes SG disassembly (Fig. [263]3c–j). While previous research has primarily focused on the role of the core SG network in SG assembly, our work shifts the perspective by revealing its equally crucial function in SG disassembly. The role of Caprin-1 in transitioning WDR45 condensates from a static gel-like state to a more dynamic liquid-like phase is essential for WDR45 localization to SGs and subsequent disassembly (Fig. [264]4c–f). This dynamic interplay between WDR45 and Caprin-1 represents a layer of regulation for SG dynamics. Our discovery underscores the importance of maintaining a dynamic balance between SG assembly and disassembly to ensure effective cellular stress responses. The protective role of WDR45 against pathological SG formation containing mutant FUS (Fig. [265]6a–d), implicated in ALS and FTD, underscores the broader relevance of our findings beyond BPAN. WDR45 depletion exacerbates the accumulation of these aberrant SGs, suggesting a potential therapeutic avenue for modulating SG dynamics in these related neurodegenerative disorders. The delayed SG recovery observed in iPSC-derived midbrain neurons from a BPAN patient further strengthens the clinical relevance of our findings and validates the in vitro observations (Fig. [266]7a, b). This patient-derived midbrain neurons model provides a powerful tool for future investigations into the molecular mechanisms linking WDR45 dysfunction to neurodegeneration. Additional studies are required to directly link these mechanisms to neuronal degeneration. Future research will focus on assessing whether restoring normal SG dynamics can mitigate neuronal degeneration in BPAN patient-derived models and mouse models. BPAN is characterized by neurodevelopmental defects in the early stage of the disease and neurodegeneration in the late stage^[267]3. However, the mechanisms underlying the transition from neurodevelopmental impairments to neurodegeneration remain unclear. Caprin-1 is essential for normal brain function, has been implicated in a neurodevelopmental disorder caused by loss-of-function variants^[268]49. Recent studies have also highlighted a strong association between SG-essential genes and neurodevelopmental disorders, further underscoring the importance of SG dynamics in brain health^[269]34. Building on these findings and our own results, we hypothesize that WDR45 functions together with Caprin-1 to regulate key processes in brain function (Fig. [270]7f), with this interaction potentially altering with aging. This dynamic interplay may provide a crucial mechanistic link between the neurodevelopmental and neurodegenerative features of BPAN, offering valuable insights into the disease progression and pathophysiology. Overall, our research sheds light on the molecular mechanisms underlying BPAN and potentially other neurodegenerative disorders associated with SG dysfunction. By elucidating the role of WDR45 in SG disassembly and its interaction with Caprin-1, we propose that targeting WDR45-Caprin-1 interactions or modulating SG dynamics could offer promising therapeutic strategies for BPAN and related neurodegenerative diseases. This approach could pave the way for developing interventions aimed at restoring normal SG dynamics and mitigating disease progression. Methods Cell culture and treatment HeLa, U2OS, HEK293T cell lines were cultured in Dulbecco’s modified Eagle medium with high glucose (HyClone, SH30022.01), and N2a was cultured in DME/F-12 (HyClone, SH30022.01), both supplementing with 10% fetal bovine serum (HUANKE, HK-CH500) and 1% penicillin–streptomycin (Vivacell, C3420-0100). Cells were maintained at 37 °C incubator supplemented with 5% CO[2]. The HEK293T (CRL-1573), HeLa (CCL-2), and Neuro-2a (CCL-131) cell lines were obtained from the American Type Culture Collection (ATCC). While U2OS cells (SCSP-5030) were purchased from Cell Bank of Chinese Academy of Sciences. iPSCs healthy control were kindly gifted from JiaYu Chen lab. All cells are regularly tested and confirmed to be free of mycoplasma contamination. For oxidative stress, cells were cultured in medium with 500 μM sodium arsenite (Millipore, 106277); for osmotic stress, cells were cultured in medium with 400 mM sorbitol (Sigma-Aldrich, S1876); for heat stress, cells were exposed to 44 °C in a humidified incubator (ESCO CelMate CO[2] INCUBATOR) with 5% CO[2]. Antibodies For Western blot, the following antibodies were used: Caprin-1 (Proteintech, 15112-1-AP, dilution: 1:500), GFP (Santa Cruz, sc-9996, dilution: 1:1000), GST (Proteintech, 10000-0-AP, dilution: 1:1000), 6*His (Proteintech, 66005-1-Ig, dilution: 1:1000), G3BP1 (Santa Cru, sc-365338, dilution: 1:1000), WDR45 (Proteintech, 19194-1-AP, dilution: 1:1000), GAPDH (Proteintech, 19194-1-AP, dilution: 1:10000), SQSTM1 (Proteintech, 18420-1-AP, dilution: 1:5000), LC3 (HUABIO, ET1701-65, dilution: 1:1000), NUP98 (HUABIO, HA722523). For immunostaining, the following antibodies were used: HA (Invitrogen, 26183, dilution: 1:500), TIAR (CST, 8509S, dilution: 1:500), Caprin-1 (Proteintech, 15112-1-AP, dilution: 1:500), Caprin-1 (Proteintech, 66352-1-Ig, dilution: 1:500), GFP (Santa Cruz, sc-9996, dilution: 1:500), G3BP1 (Proteintech, 13057-2-AP, dilution: 1:500), G3BP1 (Santa Cru, sc-365338, dilution: 1:500), WDR45 (Proteintech, 19194-1-AP, dilution: 1:400). TUJ1 (Proteintech, 66375-1-Ig, dilution: 1:400). For endo-immunoprecipitation, the following antibodies were used, G3BP1 (Santa Cru, sc-365338, dilution: 1 μg/400 μl in lysis buffer). Plasmid constructs and transfection The full length of Homo sapiens WDR45, CAPRIN1, and G3BP1 were amplified using PCR from human cDNA. The pET-MBP-WDR45 construct was generated from pET-MBP.3 C and WDR45 fragment using seamless cloning (LABLEAD, D0204P). The pLVX-WDR45-GFP construct was generated from pLVX.puro with an GFP tag inserted after gene fragment and WDR45 fragment. Constructs of GFP tag inserting before WDR45 fragment were also generated. Different truncations and mutations of WDR45 were obtained through PCR applying site-directed mutagenesis. Constructs of CAPRIN1 including pET-MBP-CAPRIN1, GST-CAPRIN1, pLVX-GFP-CAPRIN1 were generated using seamless cloning. Constructs of G3BP1 including pET-MBP-G3BP1, and pLVX-GFP-G3BP1 were generated using seamless cloning. Plasmids were transfected into corresponding cell lines using Lipofectamine 3000 (Invitrogen, L3000015) according to the manufacturer’s instructions. CRISPR–Cas9-mediated knockout cell lines Cells were plated on one well of six-well plate at a density of around 20% and transfected with 2.5 μg PX459 plasmids with corresponding guide RNA (gRNA) sequences^[271]50. gRNA sequences were designed using [272]CRISPOR (Concordet and Haeussler, 2018). Knockout plasmids were generated through T4 ligation (LABLEAD, T5205) with PX459 digested by BbsI-HF® (NEB, R3539S) and annealed oligonucleotides. Two days later, 1 μg ml^−1 puromycin (LABLEAD, P067) was added to the medium to select positive transfected cells for 3-5 days. After selection, survived cells were plated into 96 well plates at a single cell concentration. Monoclonal knockout cell lines were validated by sequencing analyses, western blot and immunostaining. The 20-nt guide sequences for the target genes are as follows: WDR45: gRNA: 5’-TGACTCAACAGCCACTTCG-3’ WDR45B: gRNA1: 5’-CATCAGTTGCACGTCTTCGA-3’ gRNA2: 5’-GAGCTGCATTGCACTCAACC-3’ CAPRIN1: gRNA1: 5’-GGAGTGAGGCGGCCGCGGGA-3’ gRNA2: 5’-TCGGGTTCCTCCGGGAGTGA-3’ Creation of stable expression cell lines Stable expression cell lines were constructed through lentivirus-mediated transfection. Briefly, 2 μg corresponding overexpression plasmids, 1 μg psPAX2 (Addgene plasmid #12260) and 1 μg pMD2.G (Addgene plasmid #12259) plasmids were used to co-transfect 293 T cells in a 3.5 cm dish firstly. Viruses were collected at 48 h post transfection and used to infect U2OS or HeLa cells for another 48 h, following selection by adding 1 μg ml^−1 puromycin to the medium for 3-5 days. Western blot was performed to validate the cell lines. siRNA transfection and RT-qPCR siRNA was transfected into cells using Lipofectamine 3000 (Invitrogen, L3000015) according to the manufacturer’s instructions. Total RNA was extracted using MagZol Reagent (Magen, R4801-01) and then reverse-transcribed (YESEN, 11141ES10). Real-time quantitative PCR was conducted with SYBR qPCR Mix (Magen, MD70101) using QuantStudio3 (Applied Bioscience). Quantitation of all target gene expression was normalized to the control gene GAPDH. qPCR primers are listed in Supplementary Table [273]2. siRNA sense sequences are listed below. si-WIPI2: 5’-GACAGUCCUUUAGCGGCA-3’ si-ATG2a: 5’- GCAUUCCCAGUUGUUGGAGUUCCUA-3’ si-ATG2b: 5’- AGGUCUCUCUUGUCUGGCAUCUUUA-3’ si-WIPI1-1: 5’- GCUGCUGGUUUGCCUAGAA-3’ si- WIPI1-2: 5’- CACCAACAGUCGACCAGAA-3’ si- WIPI1-3: 5’- GGAGAGUGUGUCUUAAUCA-3’ si- WDR45: 5’- GCAUCUAUGUGUACUCCUU-3’ si- WDR45B: 5’- GGGAUGACCUGAAGAAGAA-3’ Protein purification WDR45 (full length and mutants) and G3BP1 were subcloned to pET-MBP.3 C for protein purification. Plasmids were transformed into E. Coli BL21 competent cells. After 2 liters of E. coli were grown to OD[600] of 0.8, 0.5 M IPTG (LABLEAD, 0487) was added to induce protein expression at 16 °C for 16 hours. Pellets were collected through centrifugation and resuspended in lysis buffer (500 mM NaCl, 50 mM HEPES 7.5, 1 mM DTT, 30 mM imidazole). After sonication (60% power, 2 s on, 8 s off, working time 10 min), lysates were pelleted at 18000 g for 50 min. Supernatants were applied to the His-tag column with 5 mL beads (GenScript, L00666-100) overnight. Proteins were eluted with 300 mM imidazole in lysis buffer. The proteins were then purified by Cytiva AKTA go, collecting different fractions which were subjected to SDS-PAGE. According to the results of SDS-PAGE, certain fractions were pooled, concentrated, flash frozen in liquid nitrogen and stored at -80 °C. Western blot Cells were scraped and lysed in cold RIPA buffer (LABLEAD, R1090) for 30 min supplemented with 1% protease cocktail (LABLEAD, C0101), pipetting up and down every ten minutes, following centrifugation at 12000 g for 30 min. The pellets were discarded and supernatants were boiled in the loading buffer at 95 °C for 15 min. Protein samples were separated on an SDS-PAGE gel and transferred to a PVDF membrane (Millipore, IPVH00010), followed by incubation with primary antibodies overnight. Membrane was then washed with TBST three times, and horseradish peroxidase (HRP)-conjugated secondary antibodies (Jackson Immuno, 111-035-003) were added for visualization using ECL (LABLEAD, E1050). Protein quantification analysis was performed using NIH ImageJ (Fiji) software. Immunofluorescence and microscopy Cells were plated on glass coverslips at appropriate concentrations, followed by treatments or transfection. Cells were washed by PBS three times and fixed with 4% PFA (LABLEAD, P4500) for 15 min, followed by permeabilization with 0.2% Triton X-100 (Sigma-Aldrich, T8787) for 15 min and blocking with 3% BSA (LABLEAD, 0332-1) for 60 min. Primary antibodies were added, and incubated overnight. Corresponding Alexa Fluor Cy3, 488 secondary antibodies were added for visualization. Slides were then mounted using Mounting Medium, and antifading (with DAPI) (Solarbio, S2110). The images were captured using a Zeiss LSM880 with a ×63 oil objective. Immunoprecipitation assays Cells were seeded in 10 cm dishes at a desired concentration, following transfection or continuing to culture to a density of 90%. Cells were then scraped and lysed in 300 μL cold IP lysis buffer (25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM EDTA, 10% glycerol, 1% NP40) for 30 min, lysates were subjected to centrifugation at 12000 g for 30 min, 60 μL supernatants were boiled as input. For exogeneous immunoprecipitation, 15 μL GFP-magnetic beads (LABLEAD, GNM-50-2000) were added to the rest supernatants, following incubation overnight. For endogenous immunoprecipitation, lgG (Applygen, C1754) and primary antibodies were added to equal amounts of supernatants, following incubation overnight. Protein A/G-Sepharose beads (Thermo Scientific, 88802) were added to incubate for another 3 hours. The beads were washed 3 times before adding the loading buffer. Samples were analyzed by SDS-PAGE for immunoblotting. GST Pull-down Protein Glutathione S-transferase (GST) was conjugated to glutathione beads (GenScript, L00206-50) at 4 °C for 2 hours. GST-Caprin-1 was purified, as the process of protein purification using glutathione beads, without the step of elution. WDR45 full-length and its mutants were added to the complexes for pull-down. After incubating overnight, the beads were washed 3 times with PBS, and analyzed by SDS-PAGE using anti-GST and anti-HIS antibodies. FRAP FRAP experiments were performed on a Leica TCS-SP8 microscope with a ×63 oil immersion objective. For intracellular experiments, cells were seeded into 35 mm glass bottom dishes and were transiently transfected with WDR45-GFP or GFP-G3BP1 plasmids for 2 days. Droplets were photobleached using a laser intensity of 80% at 488 nm. Recovery was recorded for the indicated time. For in vitro experiments, Droplets of GFP-WDR45 alone or along with mCherry-Caprin-1 were photobleached using a laser intensity of 80% at 488 nm and 561 nm and recovered. Data were processed using the Fiji plugin FRAP profiler v2 and plotted with GraphPad Prism. Mass spectrometry analysis Cells stably expressed WDR45-GFP or GFP were treated with 500 μM sodium arsenite for an hour or left untreated as a control. Immunoprecipitation using anti-GFP magnetic beads was performed to track interacting proteins of WDR45. Protein samples were subjected to SDS-PAGE, and the gel blocks were cut off when samples were about to enter stacking gel. The gels were dehydrated with 200 μL of acetonitrile, air-dried for 5-10 minutes, then rehydrated and digested with trypsin (10 ng/μL). An additional 50 μL of extraction solution (60% acetonitrile, 1% TFA) was added to the gel pieces, which were sonicated for 10 minutes. The supernatant was dried by centrifugal evaporation. The resulting peptides were desalted using a Mono-spin C18 column (GL Sciences) and dissolved into 0.1% formic acid and spiked with iRT peptides before DIA analysis. LC-MS/MS analyses were performed on the Vanquish Neo UHPLC (Thermo Fisher Scientific), the trypsin digested peptides were separated using mobile phase A (0.1% formic acid (Sigma) in water) and phase B (0.1% formic acid and 80% acetonitrile (Thermo Fisher Scientific)). The Thermo Orbitrap Ascend mass spectrometer (Thermo Fisher Scientific) was configured for data-independent acquisition (DIA) by combining two experiment elements, corresponding to a full MS experiment and an MS/MS experiment, and was operated in positive polarity mode. The precursor ions were isolated by Quadrupole and fragmented by High Energy Collision Dissociation (HCD) with normalized collision energy (%) at 32. A direct DIA Analysis built in Spectronaut software (version 18.0) was used for data analysis. The MS/MS spectra were matched against the human UniProt database (20,230 entries, downloaded in August 2023). DIA files were processed in the default mode. The FDR for PSM and protein quantification were both set to 0.01. We quantified differences of protein abundance between groups using log2 fold change and applied the Student’s t-test to assess statistical significance to obtain p values. Proteins with log2 fold change > 0, and p value < 0.05 were retained. In vitro phase separation assay For single component phase separation, WDR45 Full length or mutations were diluted to different salt or protein concentrations in Eppendorf tubes and transferred to a glass slide, covered with a coverslip, and imaged with OLYMPUS IX73 equipped with 40×objectives. For co-phase separations between WDR45, G3BP1 and mCherry-Caprin-1, WDR45 was labeled with XFD488 NHS Ester (AAT Bioquest, 1812) and G3BP1 was labeled with XFD647 NHS Ester (AAT Bioquest, 1833). Proteins were diluted to appropriate concentrations as indicated under physiological conditions with 150 mM NaCl, images were captured using a Zeiss LSM880 with a ×63 oil objective. Structure Illumination microscopy (SIM) U2OS cells were seeded and grown on coverslips and treated as indicated. The mounting technique was the same as immunofluorescence. The data were collected through the Polar-SIM system (Airy Technology Co., Ltd., China). A 488 nm and 561 nm laser were used to excitation with SIM. The SIM reconstruction process was conducted using the Airy-SIM software with pre-processing (Dark) or post processing (MRA/MLE). Fluorescence intensity and Pearson correlation coefficient were analyzed using Fiji. Midbrain neurons differentiation The iPSCs were plated on matrigel (Corning® Matrigel®, 355247) and maintained in mTeSr Plus medium (STEMCELL technology, 100-0276). Midbrain neurons were induced using DAXING technology neuron induction kits according to the manufacturer’s recommendations. Briefly, 4×10^5 iPSCs were plated on neuron coating matrix (DAXIANG, NK100107) and kept in mTeSr Plus. The next day, medium was changed to Neuron progenitor induction medium (DAXIANG, NK100101) for 14 days. Subculturing was necessary when cell confluency reached 90%. After 14 days, neuron progenitor cells were successfully induced and maintained in NPC expansion medium (DAXIANG, ND100102). 2×10^5 neuron progenitor cells were plated in Midbrain Differentiation medium (DAXIANG, NK100103) for 14 days and Midbrain Maturation medium (DAXIANG, NK100107) for 7 days, changing medium daily. For oxidative treatment, cells were exposed to 500 μM sodium arsenite for 30 min, for heat shock, cells were exposed to 44 °C for 1 hour. Living cell imaging HeLa knock in G3BP1-GFP cell line was used for living cell imaging. Briefly, cells growing on confocal dish (Cellvis, D35C4-20-1.5-N) were transfected with siRNA targeting WDR45 or control, then following incubation for 48 h. For arsenite (500 μM) stress phase, images were captured at 15 min, 40 min, 60 min. Using Nikon Elements software with definite focus engaged, multipoint images were taken every 20 s with the 488-nm laser at 10% power, average is 2, zoom is 1024*1024. For recovery phase (using fresh culture medium after arsenite stress (500 μM, 60 min)), images were captured at 0 min, 60 min, 120 min and 150 min. Multipoint images were taken every 1 min with the 488-nm laser at 5% power, average is 2, zoom is 1024*1024. The living cell images were collected by Nikon AXR NSPARC and analyzed by Nikon Elements viewer and image J software. The LUTS was changed in the same situation. Cells were maintained at 37 °C incubator supplemented with 5% CO[2]. Statistics & reproducibility For the immunostaining of treated cells, cells harboring SGs were quantified manually, at least three fields per experiment were analyzed; areas of FUS-positive granules were measured using Fiji, and all cells were considered for the quantification. Quantification of western strips was performed using Fiji software, and at least three biological repeats were measured. Colocalization analysis was performed using the Fiji plugin Coloc2. GraphPad Prism was used for statistical analysis. Statistical values are displayed as the means ± SD (standard deviation)/SEM (standard error of the mean). p > 0.05 was considered not significant. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 by Student’s t-test, one-way ANOVA, or two-way ANOVA. The details are included in the corresponding figure legends. All experiments were repeated at least three times with similar results. Reporting summary Further information on research design is available in the [274]Nature Portfolio Reporting Summary linked to this article. Supplementary information [275]Supplementary Information^ (10.6MB, pdf) [276]DOASF^ (45.4KB, pdf) [277]Supplementary Movie 1^ (9.7MB, mp4) [278]Supplementary Movie 2^ (9.7MB, mp4) [279]Supplementary Movie 3^ (78.8MB, mp4) [280]Supplementary Movie 4^ (86.9MB, mp4) [281]Reporting summary^ (2.8MB, pdf) [282]Transparent Peer Review file^ (959.1KB, pdf) Source data [283]source data^ (19.8MB, xlsx) Acknowledgements