Introduction

Esophageal squamous cell carcinoma (ESCC), with a 5-year survival rate of approximately 15% to 20%, is one of the most lethal gastrointestinal malignancies worldwide [1,2,3]. Although some targeted anticancer drugs, such as gefitinib and PD-L1 blockers, have been adopted in the clinic [4], the mortality rate of ESCC is still relatively high owing to invasion and distant metastasis. Large-scale whole-exome sequencing (WES) of ESCC has identified some high-frequency gene mutations, including copy number alterations and somatic mutations [5,6,7]. In addition, epigenetic alterations in ESCC, such as DNA methylation and histone acetylation, have also been partially characterized [8]. Despite great advances in the genomic and epigenetic aspects of ESCC, the underlying mechanisms of tumor progression remain poorly understood. Hence, it is imperative to further investigate the mechanisms.

Purine-rich element binding protein alpha (PURα), encoded by PURΑ, is a single-stranded DNA/RNA-binding protein that is highly conserved from bacteria to humans [9]. PURΑ knockout in mice and PURΑ mutation in humans both result in severe neurological disease [10,11,12,13,14]. Abnormal PURα expression is also involved in the progression of several cancers, such as acute myeloid leukemia (AML) and prostate cancer [15, 16], and our previous results have shown that overexpression of PURα promotes ESCC progression [17]. Thus far, PURα has been implicated primarily in DNA replication, transcription and the cell cycle [18,19,20,21,22,23]. Recent reports have indicated that PURα in the cytoplasm encapsulates specific RNAs with some RNA-binding proteins to regulate mRNA transport [24,25,26,27,28], and emerging evidence suggests that PURα is a novel component of cytoplasmic stress granules [25, 29,30,31]. Stress granules are cytoplasmic RNA–protein complexes that form when translation initiation is limited [32, 33] and have been proposed to play an important role in neurodegenerative diseases and tumor progression [34, 35]. For example, PURα colocalized with mutant FUS in stress granules to modulate amyotrophic lateral sclerosis (ALS) pathology [25, 31]. In addition, PURα is known to associate with noncoding RNAs (e.g., TAR RNA [36], BC200 [26] and circSamd4 [37]). PURα strongly influences the development and progression of disease by regulating DNA replication, transcription and mRNA transport, but whether PURα participates in tumor progression by regulating mRNA-based processes remains unclear.

Here, we found that PURα participates in the formation of cytoplasmic stress granules and that the expression level of cytoplasmic PURα was significantly increased in ESCC tissues compared to nontumorous tissues and that ESCC patients with high expression levels of cytoplasmic PURα had a lower survival rate than those with low expression levels. We further revealed that PURα repressed the mRNA translation initiation of insulin-like growth factor binding protein 3 (IGFBP3) by forming cytoplasmic stress granules. In addition, knockdown of IGFBP3 significantly reversed the inhibitory effects of PURα loss on the cell proliferation, migration and invasion properties of KYSE170 ESCC cells. In brief, our results support that cytoplasmic PURα mediates ESCC progression by binding to the mRNA 3´UTR.

Results

Cytoplasmic PURα participates in the formation of stress granules and significantly correlates with ESCC progression

It has been commonly reported that PURα is involved in the progression of several cancers as a transcription factor [9, 18, 23]. Intriguingly, immunofluorescence staining indicated that there was the considerable cytoplasmic localization of PURα in ESCC cells and that cytoplasmic PURα was evenly dispersed as granules or accumulated around the nucleus in nongranules (Fig. 1A). Increasing evidence has reported that PURα is a core component of cytoplasmic stress granules [25, 29,30,31], suggesting that cytoplasmic PURα-positive granules in ESCC cells are likely a form of stress granules. To this end, the colocalization between PURα-positive granules and G3BP1, a well-known cytoplasmic stress granule maker [32, 33, 38], was further detected by immunofluorescence staining. It was observed that G3BP1 is localized in PURα-positive granules and that the number of PURα/G3BP1-positive granules under stress conditions markedly increased compared with those under native conditions, while the number of G3BP1-positive granules markedly decreased after the loss of PURα (Fig. 1B, C), demonstrating that cytoplasmic PURα in ESCC cells participates in the formation of stress granules. In addition, we also observed that there was more expression of PURα in the cytoplasm than in the nucleus in the ESCC tissues by immunohistochemical staining (n = 526) (Fig. 1D, E), and the protein fractionation analysis also indicated that PURα was additionally localized in the cytoplasm to a much greater extent than in the nucleus of ESCC cells (Fig. S1A). Furthermore, we compared the expression of cytoplasmic PURα in ESCC tissues (n = 282) and adjacent nontumorous epithelia (n = 282) and observed that cytoplasmic PURα expression was significantly increased in ESCC tissues (Fig. 1D, F). Importantly, Kaplan–Meier survival analysis of a total of 526 ESCC patients showed that ESCC patients with high expression levels of cytoplasmic PURα had a lower survival rate than those with low PURα (Fig. 1G), implying that cytoplasmic PURα is linked to ESCC progression. There were no correlations between PURα levels and sex, age, tumor differentiation or other factors (Supplementary Table S1).

Fig. 1: Cytoplasmic PURα participates in the formation of stress granules and significantly correlates with ESCC progression.
figure 1

A The localization of endogenous PURα in esophageal epithelium Het-1A and ESCC cancer (KYSE170) cells was visualized by immunofluorescence assay. PURα proteins dispersed in the cytoplasm as granules in KYSE170 cells or accumulated around the nucleus in Het1A cells. Scale bars: 30 μm. B The colocalization between endogenous PURα and the stress granule maker G3BP1 was visualized in wild-type (WT) and PURα-deficient KYSE170 (KO) cells by immunofluorescence staining. Scale bars: 30 μm. C The number of PURα/G3BP1-positive granules under stress conditions or not was calculated separately. ***p < 0.001; ns, not significant. D PURα expression in ESCC tumor tissues (first and second panels) and adjacent nontumor epithelia (third panel) was compared by immunohistochemical staining (IHC). PURα protein mainly located in cytoplasm (first panel) or nucleus (second panel) is shown. The representative region (black frame) at low magnification (40×, left) was amplified at high magnification (100×, right). Scale bars: 50 μm. E Violin plots of the statistical data regarding the IHC score of PURα protein in the cytoplasm and nucleus of tumor tissues (n = 526). ***p < 0.001 by Mann–Whitney test. F Violin plots of the statistical data regarding the IHC scores for cytoplasmic PURα in ESCC (tumor) and adjacent nontumor (normal) tissues (n = 282) were drawn. ***p < 0.001 by Mann–Whitney. G Kaplan–Meier analyses of overall survival. Patients with high cytoplasmic PURα expression (n = 296) had a significantly lower overall survival rate than patients with low cytoplasmic PURα expression (n = 230).

Extensive RNA targets of PURα in ESCC cells were revealed by CLIP-seq analysis

Cytoplasmic stress granules are ribonucleoprotein granules and are involved in the regulation of RNA homeostasis [32, 33], so we speculated that cytoplasmic PURα likely modulates ESCC progression through interaction with mRNA. Cytoplasmic PURα notably affects brain development in humans and mice as an RNA-binding protein, but the characteristics of RNA bound to PURα have not been fully elucidated [10,11,12,13,14, 24, 26,27,28]. We thus first identified RNA targets of human PURα in ESCC KYSE510 cells by CLIP-seq analysis according to reported methods [39]. In total, high-throughput sequencing yielded ~38.5 and ~34.4 million raw reads from two independent replicated PURα CLIP-seq datasets (Supplementary Table S2). After discarding low-quality raw reads and normalization, ~17.0 and ~15.2 million clean reads were generated, respectively. Of these clean reads, 82.27% (~13.9 million) and 83.52% (~12.7 million) were unambiguously mapped to the human reference genome (hg38) (Supplementary Table S3). The vast majority of uniquely mapped reads (62.98% and 57.48%) mapped to introns. Then, PURα-binding sites were predicted with a peak calling algorithm as previously reported [

Fig. 7: Schematic of the mechanism by which cytoplasmic PURα inhibits mRNA translation to mediate ESCC progression.
figure 7

As a novel component of cytoplasmic stress granules, PURα preferentially binds to UG-/U-rich motifs located in the 3’UTRs of mRNAs and recruits translation initiation factors to attenuate mRNA translation.