Graphical abstract graphic file with name fx1.jpg [55]Open in a new tab Highlights * • ALDOA overexpression in various retinoblastoma cell types and tissues * • ALDOA knockdown reduces retinoblastoma cell viability and alters metabolism * • Itaconate inhibits retinoblastoma proliferation in both in vitro and in vivo models * • Targeting ALDOA holds potential for advancing retinoblastoma therapies __________________________________________________________________ Molecular biology; Cancer; Transcriptomics Introduction Retinoblastoma (RB) is the most prevalent ocular malignancy in infants and young children. It constitutes 2–4% of all pediatric tumors, wielding a profound impact on both the visual acuity and life expectancy of affected individuals.[56]^1 The incidence of RB typically ranges from 1 in 20,000 to 1 in 15,000, yielding approximately 8,000–9,000 new cases among children annually.[57]^2 Among the RB patients who experience metastasis, an alarming 80% eventually face a fatal outcome, with the invasive extraocular extension emerging as pivotal contributors to the metastatic process.[58]^3 Recent years have witnessed notable progress in the diagnosis and management of RB, leading to substantial improvements in both survival and rates of eye preservation. However, it is crucial to note that the advanced RB remain refractory to current therapeutic modalities, resulting in a less favorable prognosis. The phenomenon, known as aerobic glycolysis or the Warburg effect, has evolved beyond a mere adaptation to the environment, now recognized as an integral component of malignant phenotypes and tumor metabolic reprogramming.[59]^4 Enhanced glycolysis in tumors not only fuels their proliferation but also provides intermediates for synthesizing critical biomolecules like nucleotides, lipids, and proteins.[60]^5 Consequently, targeting tumor glycolysis has arisen as a promising strategy in cancer therapy, as tumor cells adjust their energy metabolism to meet the demands of growth, biosynthesis, and redox reactions. Aldolase is a pivotal enzyme within the glycolysis pathway, facilitating the breakdown of 1,6-diphosphate-D-fructose into 3-phosphate-D-glyceraldehyde, and subsequently interacting with α-dihydroxyacetone phosphate.[61]^6 The aldolase family comprises three members: ALDOA, ALDOB, and ALDOC. Notably, ALDOA has been observed to be overexpressed in a variety of malignant tumors, including gastric cancer, hepatocellular carcinoma, colorectal cancer, and cervical cancer.[62]^7^,[63]^8^,[64]^9^,[65]^10 Our recent research into the metabolic microenvironment of RB has revealed an upregulation of the glycolytic pathway.[66]^11 In this study, we aim to investigate the expression of key glycolytic enzymes in RB at the single-cell level. By integrating single-cell RNA sequencing (scRNA-seq) datasets from RB and normal retinal tissues, we compared gene expression profiles and confirmed the upregulation of ALDOA in RB. Subsequent in vitro and in vivo experiments demonstrated the effectiveness of targeted ALDOA inhibition in suppressing RB growth. Results Elevated ALDOA expression in retinoblastoma GSEA of DEGs between retina and RB revealed significant enrichment in gene sets related to amide biosynthesis, glycolysis, catalytic activity/metabolism, and ATP synthesis ([67]Figure 1A). This suggests a significant alteration in energy metabolism in RB, highlighting the potential crucial role of the glycolytic pathway in RB. Expression patterns of key genes within the glycolytic pathway was showed in heatmap ([68]Figure 1B). Through dimensionality reduction and subsequent clustering analysis utilizing Uniform Manifold Approximation and Projection (UMAP), we visualized the expression profiles of Aldolase A (ALDOA) in RB and normal retina ([69]Figure 2A). The analysis unveiled a high expression level of ALDOA in RB ([70]Figure 2B), particularly elevated in extraocular RB compared to intraocular cases ([71]Figure 2C). Branched expression analysis modeling unveiled a clear upregulation of ALDOA expression in retinoma-like cells as they progressed through pseudotime. However, this unique expression profile was not evident in other cell types ([72]Figure S1). To validate ALDOA expression in RB, we examined its levels in human RB tissue samples and RB cell lines (WERI-RB1, Y79). The results demonstrated upregulated ALDOA mRNA and protein levels in RB cells compared to ARPE-19 ([73]Figures 3A and 3B). Additionally, we analyzed ALDOA mRNA expression using the [74]GSE125903 dataset[75]^12 from the Gene Expression Omnibus (GEO) database, distinguishing between invasive and non-invasive RB cases. The analysis revealed higher ALDOA expression in invasive RB cases compared to non-invasive ones ([76]Figure 3C). Proteins extracted from tissues of 5 intraocular and 5 extraocular RB patients revealed higher ALDOA protein expression in extraocular RB, as detected by western blotting ([77]Figure 3D). IHC results further confirmed significantly higher ALDOA expression in extraocular RB compared to intraocular RB and normal retina ([78]Figure 3E). Clinicopathological analysis revealed a trend toward higher ALDOA expression in cases with high-risk features like scleral and optic nerve invasion. Notably, ALDOA expression was significantly lower in younger patients (<2 years old) ([79]Figure S2). Figure 1. [80]Figure 1 [81]Open in a new tab Enrichment analysis and glycolytic pathway expression in retinoblastoma (A) Gene set enrichment analysis (GSEA) of differentially expressed genes (DEGs) between normal retina and retinoblastoma (RB) samples. (B) Heatmap representation of the expression patterns of key genes within the glycolytic pathway, highlighting their differential expression in RB compared to normal retina. Figure 2. [82]Figure 2 [83]Open in a new tab Visualization of ALDOA expression profiles in retinoblastoma and normal retina (A) Visualization of the expression profiles of Aldolase A (ALDOA) in retinoblastoma (RB) and normal retina using uniform manifold approximation and projection (UMAP). (B) Comparison of ALDOA expression between RB and normal retina. (C) Comparison of ALDOA expression between extraocular RB and intraocular case. (D) ALDOA expression across various cell types. Figure 3. [84]Figure 3 [85]Open in a new tab ALDOA overexpression in various retinoblastoma cell types and tissues (A and B) Quantitative PCR (A) and western blot assays (B) revealed significantly higher messenger RNA (mRNA) and protein levels of ALDOA in retinoblastoma (RB) cells compared to normal cells. (C) ALDOA mRNA expression levels in non-invasive and invasive retinoblastoma cases from the [86]GSE125903 dataset. (D) Western blot analysis of ALDOA protein expression in tissues obtained from 5 intraocular and 5 extraocular RB patients, demonstrating elevated ALDOA protein levels in extraocular RB. (E) Immunohistochemistry (IHC) staining and a statistical analysis of ALDOA expression in normal retina, intraocular RB tissue, and extraocular RB tissue. R, retina; RB, retinoblastoma; S, sclera; VC, vitreous cavity; ON, optic nerve. Data are presented as means ± standard deviations. Statistical significance was determined using Student’s t test, ∗p < 0.05, ∗∗p < 0.001, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001. Disruption of retinoblastoma cell growth and energy metabolism by ALDOA downregulation Given the observed ALDOA overexpression in RB, we investigated the functional consequences of ALDOA downregulation in RB cell lines. The qRT-PCR and western blotting results showed a decrease in ALDOA mRNA and protein expression levels after transfection ([87]Figures 4A and 4B). The CCK-8 assay indicated that upon ALDOA knockdown in Y79 and WERI-RB1 cells, noticeable alterations in cell viability were observed from the second day, with more pronounced differences becoming evident by the third day when compared to the si-NC groups ([88]Figure 4C). Additionally, the EdU assay results showed a decrease in the proliferation ability of Y79 and WERI-RB1 cells following ALDOA knockdown ([89]Figure S3). The colony formation assays revealed a significant decrease in RB cells colony formation abilities following the knockdown of ALDOA ([90]Figure 4D). Lactate assay results demonstrated a marked reduction in extracellular lactate level in RB cells following ALDOA knockdown ([91]Figure 4E), accompanied by elevated levels of reactive oxygen species and decreased ATP levels ([92]Figures 4F and 4G). Figure 4. [93]Figure 4 [94]Open in a new tab Functional implications of ALDOA downregulation in retinoblastoma cell lines (A and B) Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) (A) and western blotting analysis (B) revealed a substantial reduction in ALDOA mRNA and protein expression levels following transfection of RB cell lines. (C) The CCK-8 assay demonstrated inhibited cell proliferation in response to ALDOA downregulation. (D) Colony formation experiments indicated a reduced tumorigenic potential of RB cell lines after ALDOA knockdown. (E) Extracellular lactate analysis exhibited a notable decrease in lactate levels in RB cells treated with siALDOA for 48 h compared to the negative control. (F) Levels of reactive oxygen species (ROS) in RB cells treated with siALDOA for 48 h were increased compared to the negative control. (G) Intracellular ATP levels in RB cells treated with siALDOA for 48 h were reduced in comparison to the negative control. Data are presented as means ± standard deviations. Statistical significance was determined using Student’s t test, with ∗p < 0.05, ∗∗p < 0.001, and ∗∗∗p < 0.001 denoting significance. Signaling alteration in response to ALDOA downregulation in retinoblastoma Next, we performed transcriptome sequencing on both the si-ALDOA and si-NC WERI-RB1 to explore the downstream alterations resulting from ALDOA downregulation. Our transcriptome analysis revealed significant changes in gene expression patterns, highlighting 162 upregulated genes including PHRF1, SUSD2, IGFN1, UNC79, and 136 downregulated genes including PTP4A3, ARHGAP27, MYT1, CLK2 ([95]Figures 5A and 5B). Further pathway enrichment analysis of these differentially expressed genes (DEGs) indicated potential shifts in biological functions in RB following ALDOA downregulation. Significantly, the upregulated DEGs were notably enriched in pathways related to “Organic acid transport”, “Amino acid transport”, and “Carboxylic acid transport” ([96]Figure 5C). Conversely, the downregulated DEGs were predominantly associated with pathways involving “DNA-binding transcription activator activity” and “histone methyltransferase activity” ([97]Figure 5D). Gene set enrichment analysis (GSEA) further revealed that after ALDOA knockdown, upregulated DEGs were enriched in pathways such as glycosphingolipid biosynthesis, while downregulated DEGs were enriched in metabolic pathways like oxidative phosphorylation, glutathione metabolism, and fatty acid metabolism ([98]Figure S4). To validate the expression levels of the identified DEGs, we conducted qRT-PCR on selected genes displaying significant expression differences. The results demonstrated a substantial upregulation of SUSD2, ARHGAP27, and CLK2 mRNA levels in RB cell lines compared to the ARPE-19 ([99]Figure 5E). Furthermore, in RB cell lines where ALDOA was knocked down, these genes exhibited altered mRNA expression patterns, with increased expression of SUSD2 and decreased expression of ARHGAP27 and CLK2 ([100]Figure 5F). Western blot results showed that after ALDOA knockdown, the protein level of SUSD2 increased while the levels of CLK2 and ARHGAP27 decreased in WERI-RB1 cells. However, in Y79 cell lines, an increase in SUSD2 protein level was not detected after ALDOA knockdown, and the trends for the other two proteins were also decreasing ([101]Figure 5G). Furthermore, to individually knock down SUSD2, ARHGAP27, and CLK2, three pairs of siRNAs were designed for each gene ([102]Figure 5H). CCK8 experiments revealed that knocking down ARHGAP27 led to a decrease in the proliferation capacity of RB cells ([103]Figure 5I). Figure 5. [104]Figure 5 [105]Open in a new tab Effects of ALDOA downregulation on WERI-RB1 cell line (A) RNA-seq analysis was employed to examine alterations in the mRNA expression profile of WERI-RB1 cells following ALDOA downregulation. (B) Heatmap analysis illustrating differentially expressed genes (DEGs) in WERI-RB1 cells subjected to ALDOA knockdown. (C and D) Pathway enrichment analysis focused on the upregulated DEGs (C) and downregulated DEGs (D). Upregulated DEGs were significantly associated with pathways related to “Organic acid transport”, “Amino acid transport”, and “Carboxylic acid transport”, while downregulated DEGs were primarily linked to “DNA-binding transcription activator activity” and “Histone methyltransferase activity”. (E) Quantitative PCR analysis of selected DEGs (SUSD2, ARHGAP27, and CLK2). (F) Quantitative PCR analysis of SUSD2, ARHGAP27, and CLK2 in RB cell lines with ALDOA knockdown. (G) Western blot analysis showing changes in SUSD2, ARHGAP27, and CLK2 protein levels following ALDOA knockdown. (H) Western blot analysis of SUSD2, ARHGAP27, and CLK2 protein knockdown. (I) Proliferation changes in WERI-RB1 cells following knockdown of SUSD2, ARHGAP27, and CLK2. Statistical significance was assessed using Student’s t test, with ∗p < 0.05 and ∗∗p < 0.001 indicating significance. Itaconate suppresses retinoblastoma cell proliferation and inhibits WERI-RB1 xenograft growth Itaconate is an enzymatic inhibitor of ALDOA, which suppresses the catalytic activity of ALDOA in the glycolytic pathway through covalent modification of Cys73 and Cys339, without altering the protein expression level of ALDOA.[106]^13 Hence, we utilized itaconate as a means to inhibit the enzymatic activity of ALDOA, while keeping its protein expression level unaltered ([107]Figure 6A). Results from the CCK8 assay demonstrate a reduction in RB cell viability upon the addition of itaconate ([108]Figure 6B). Moreover, the introduction of itaconate led to alterations in energy metabolism within RB cells. Mass spectrometry analysis revealed alterations in cellular metabolite levels upon treatment with itaconate in RB cell lines. Specifically, there was a downward trend in the levels of metabolites involved in glycolysis pathways such as 3-Phospho-D-glycerate, Beta-D-Fructose 6 phosphate, and D-Glucose-6-phosphate, although not statistically significant ([109]Figure 6F). However, metabolites associated with oxidative phosphorylation pathways including L-Malate acid, Fumarate, and Oxaloacetate exhibited a significant decrease ([110]Figure 6G). To further investigate, an RB murine model was established by injecting WERI-RB1 cells into the eyes of nude mice. 14 days post-grafting, the animals were randomized into groups and received intraocular administrations of either PBS, itaconate, or melphalan, with enucleation performed on day 40 ([111]Figures 7A and 7B). Analysis of eye weight revealed that both the itaconate and melphalan treatment groups had lower eye weights compared to the control group, although these differences were not statistically significant ([112]Figure 7C). External examination of the eyes and H&E staining showed significant corneal neovascularization and extraocular invasion in the PBS group, with extensive tumor cell infiltration in the vitreous body and anterior chamber. In contrast, the itaconate treatment group exhibited tumor cells primarily located anterior to the retina. The melphalan-treated mice had fewer intraocular tumor cells but showed signs of ophthalmatrophia and retinal structural abnormalities ([113]Figure 7D). Histological analysis revealed ophthalmatrophia in 40% of the melphalan-treated eyes and 20% of the itaconate-treated eyes, while extraocular invasion was observed in 30% of the PBS group and 10% of the itaconate group ([114]Figure 7E). TUNEL staining showed a higher incidence of TUNEL-positive cells in the tumors of the itaconate group compared to PBS controls ([115]Figure 7F). Additionally, the itaconate group had a lower proportion of TUNEL-positive cells in the retina compared to the melphalan-treated group ([116]Figure 7G). These observations indicate that itaconate is effective in targeting intraocular RB cells similarly to melphalan, while maintaining a more favorable safety profile for retinal tissue. Figure 6. [117]Figure 6 [118]Open in a new tab Itaconate-mediated inhibition of ALDOA and its impact on retinoblastoma in vitro and in vivo (A) Western blotting analysis Itaconate serves as an enzymatic inhibitor of ALDOA while preserving ALDOA’s protein expression level. (B) CCK8 assay reveal a reduction in RB cell viability following the addition of itaconate. (C, D, and E) Introduction of itaconate induces alterations in energy metabolism within RB cells, leading to reduced lactate production (C), increased generation of reactive oxygen species (ROS) (D), and decreased ATP generation (E). (F) Metabolite changes in the glycolytic pathway in RB cells treated with itaconate. (G) Metabolite changes in the oxidative phosphorylation pathway in RB cells treated with itaconate. Statistical significance was assessed using Student’s t test, with ∗p < 0.05 and ∗∗p < 0.001 indicating significance. Figure 7. [119]Figure 7 [120]Open in a new tab Itaconate treatment in an RB murine model (A) Study schematic illustrating the experimental design. (B) Gross appearance of eyeballs 40 days post-xenograft. (C) Eye weight analysis (n = 10). (D) Representative views of enucleated eyes and H&E staining from each treatment group. (E) Proportion of eyes with ophthalmatrophia and extraocular RB in the different treatment groups. (F) Representative TUNEL images and quantitative analysis of intratumoral cells comparing the itaconate and melphalan groups. (G) Representative TUNEL images and quantitative analysis of retinal cells comparing the itaconate and PBS groups. Data are presented as means ± SD. Statistical significance was determined by unpaired, two-tailed Student’s t test, with ∗p < 0.05 and ∗∗p < 0.01 indicating significance. Discussion In this study, we integrated the scRNA-seq datasets of normal retinal tissue and RB tissue, uncovering a significant upregulation of the glycolytic enzyme ALDOA in tumor tissue. Additionally, GSEA identified enrichment of gene sets containing ALDOA related to RB, particularly those associated with ATP generation, protein synthesis, and glycolysis. Next, we confirmed the elevated expression of ALDOA in both cell lines and patient tissues, and noted a significant increase in extraocular RB. Through the application of siRNA interference and the ALDOA inhibitor itaconate, we demonstrated that ALDOA inhibition alter tumor energy and metabolism and efficiently diminished the viability of RB cells and the tumor progression in the RB mouse model. Furthermore, by using transcriptome sequencing, we uncovered possible downstream targets under the control of ALDOA, such as SUSD2, ARHGAP27, and CLK2. The mutation or loss of RB1 is a fundamental etiological factor in the development of RB. RB1 encodes the RB protein, which directs cellular metabolism reprogramming through E2F-induced transcriptional mechanisms, involving processes such as glucose oxidation phosphorylation, fatty acid oxidation, and amino acid synthesis.[121]^14 Nonetheless, the exact role of metabolic reprogramming in tumor progression and the specific metabolic enzymes implicated in the development of RB remain incompletely understood. ALDOA, a member of the aldolase enzyme family, has been the subject of extensive research, shedding light on its intricate interactions with hypoxia-inducible factor-1 (HIF-1) and its crucial role in modulating the glycolytic pathway and the process of epithelial-mesenchymal transition (EMT).[122]^15 It is worth noting that ALDOA expression levels have been thoroughly examined across various malignancies, revealing a direct link between ALDOA expression and the aggressiveness and invasiveness of tumors. Furthermore, comprehensive survival analyses consistently emphasize a strong positive correlation between increased ALDOA expression and an adverse prognosis.[123]^16 In line with previous findings, our research results validate a noticeable upregulation of ALDOA in RB tissue. Importantly, in cases of extraocular RB, ALDOA demonstrates higher expression levels compared to intraocular cases, highlighting the pivotal role of ALDOA in promoting the initiation and progression of RB. Recent research has increasingly focused on RB invasiveness, identifying targets such as UBE2C, SOX4, and the MCM family.[124]^17^,[125]^18^,[126]^19 However, additional investigation is needed to clarify the specific role of ALDOA in the extraocular invasion of RB. Liquid biopsies, especially aqueous humor biopsies, show promise for predicting RB prognosis and assessing chemotherapy efficacy.[127]^20^,[128]^21^,[129]^22^,[130]^23 Investigations utilizing aqueous humor biopsy have shown potential in aiding RB prognosis and assessing chemotherapy efficacy.[131]^24^,[132]^25 Thus, further exploration of tumor-related glycolytic enzymes in aqueous humor or blood samples may provide additional support for the diagnostic significance of ALDOA in extraocular RB. In the course of RB development, distinct cell types play varying roles. Our analysis of scRNA-seq dataset reveals a pronounced upregulation of ALDOA in specific cell types, particularly cone cell precursors, RB-like cells, and retinoma like cells. Cone precursor like cells represent an early-stage cell population with the inherent potential for differentiation into cones or malignant cells.[133]^26 RB-like cells embody a highly proliferative malignant cell population characterized by a reduction in photoreceptor characteristics.[134]^27 Retinoma like cells serve as transitional cell stages between premalignant cone precursors and fully developed tumor cells.[135]^28 The specific high expression of ALDOA within these three cell types suggests its potential oncogenic role in RB. Apart from tumor cells, the tumor microenvironment comprises a complex network of cells, stroma, blood vessels, and immune cells surrounding the tumor.[136]^29^,[137]^30 Within the tumor microenvironment, the expression levels of ALDOA may be regulated, and ALDOA can influence the metabolism and survival status of tumor cells. Specifically, ALDOA may influence the function and status of other cells within the tumor microenvironment by affecting the energy metabolism and signaling pathways of tumor cells.[138]^31^,[139]^32 However, in this study, the impact of ALDOA on other cells within the RB microenvironment, such as immune cells, stromal cells, and endothelial cells, has not yet been investigated. Additionally, ALDOA may modulate factors such as oxygen concentration, pH balance, and nutrient supply within the tumor microenvironment, affecting the growth and chemoresistance of tumor cells.[140]^33^,[141]^34^,[142]^35 Moreover, previous studies have implicated the POU2F1-ALDOA axis in promoting tumor cell proliferation and chemoresistance.[143]^36 In this study, RNA sequencing revealed a significant alteration in POU2F1 expression following ALDOA knockdown, suggesting that the POU2F1-ALDOA axis may play a critical regulatory role in RB. Itaconate has the capability to inhibit the catalytic activity of ALDOA in the glycolytic pathway without altering the expression level of the ALDOA protein. Initially discovered as a metabolite activated in macrophages, itaconate plays a critical role in the negative regulation of inflammatory responses by suppressing the production of inflammatory factors associated with macrophage activation.[144]^37 This regulatory process primarily functions through the alkylation of KEAP1, leading to the activation of NRF2.[145]^38 In the research of tumor, there has been a hypothesis that itaconate might indirectly influence tumor progression by its impact on macrophages.[146]^39 However, it remains uncertain whether itaconate directly regulates tumor progression. In this study, we found that although the intravitreal injection of itaconate did not result in a reduction in the rate of intraocular tumorigenesis, there was a notable decrease in the size of intraocular tumors, as well as a reduction in structural damage to the eyeball following the treatment. These observations suggest the potential of itaconate as an adjunctive therapy for RB. One of the primary challenges in translating preclinical findings into clinical applications is navigating the complex regulatory landscape. For itaconate to be considered for clinical use in treating RB or other tumors, detailed pharmacokinetic studies must be conducted. Additionally, thorough investigation of potential side effects is necessary to ensure patient safety. Genes such as PHRF1, SUSD2, IGFN1, and UNC79 were identified as the top upregulated genes in ALDOA knockdown samples. SUSD2, situated on chromosome 22, encodes a transmembrane protein that plays a crucial role in cell-cell interactions and cell-matrix adhesion. Studies have indicated that SUSD2 acts as a tumor suppressor in RB by regulating the occurrence, development, and angiogenesis of RB, making it a potential target for RB treatment.[147]^40 Conversely, genes including PTP4A3, ARHGAP27, MYT1, and CLK2 were identified as the top downregulated genes in ALDOA knockdown samples. ARHGAP27 functions as a Rho GTPase-activating protein and is associated with pediatric chronic myeloid leukemia as well as ovarian cancer.[148]^41^,[149]^42 CLK2, a phosphorylation-involved splicing protein kinase, exerts a promotional role in various cancers.[150]^43^,[151]^44 These differentially expressed genes might play important roles in RB progression, part of which were validated in this study, while the intrinsic interaction network in RB still requires further exploration. In conclusion, our study illuminates the important role of ALDOA in driving tumor growth in RB. These findings not only offer insights into the metabolic dynamics of RB but also present potential target candidates for the diagnosis and therapy of RB patients. Limitations of the study Our study has certain limitations that warrant discussion. First, the relatively small number of clinical samples used may constrain the analysis, potentially limiting the generalizability of our findings. Expanding the sample size and diversifying patient demographics would strengthen the robustness of our conclusions. Second, incorporating primary cells into future investigations could better capture the heterogeneity of RB, thus enhancing the validity of our results. Additionally, further exploration into patient outcomes and correlation with relevant clinical parameters would provide deeper insights into the clinical significance of our findings. Lastly, while our study successfully identifies downstream targets, a more comprehensive investigation is needed to elucidate the mechanistic intricacies of how ALDOA influences these targets and the broader signaling pathways involved. Resource availability Lead contact Further information and requests for resources should be directed to and will be fulfilled by the lead contact, Rong Lu (lurong@gzzoc.com). Materials availability This study did not generate new unique reagents. Data and code availability * • The raw sequence data reported in this paper have been deposited in the Genome Sequence Archive in National Genomics Data Center, China National Center for Bioinformation/Beijing Institute of Genomics, Chinese Academy of Sciences (GSA-Human: HRA008175) that are publicly accessible at [152]https://ngdc.cncb.ac.cn/gsa-human. Original western blot images have been provided in the supplementary material. * • This paper does not report original code. * • Any additional information required to reanalyze the data reported in this paper is available from the [153]lead contact upon request. Acknowledgments