PIWIL1 is Upregulated in primary tumor-derived samples from Colorectal Cancer Patients Compared to Healthy Individuals.
Data mining was performed to determine the expression of the four PIWI proteins codified by the human genome, in samples-derived from patients with CRC. PIWIL1, PIWIL2, PIWIL3 and PIWIL4 RNA-seq data from colon primary tumor and normal cohorts-derived samples were analyzed using the UCSC Xena Browser (Goldman et al., 2020), integrating data from TCGA, TARGET and GTEX databases. RNA-seq data from 596 samples were ranked according to PIWI gene expression levels, revealing a positive correlation between PIWIL1 and tumor status (Fig. 1A-C).
PIWIL1 is localized in the nucleus in colon cancer cells.
Cell lines derived from human colon carcinoma and the beningn colon epithelial cell line (CCDCoN) were assessed for PIWIL1 expression at transcript and protein level. All cell lines expressed variable levels of PIWIL1 transcripts, measured by RT-PCR, and protein levels assesed by immunoprecipitation followed by Western blot (Figs. 2A and 2B, respectively). Indirect immunofluorescence (IF) to determine PIWIL1 subcellular localization showed a predominant nuclear localization in Caco-2 (Fig. 2C) and other CRC cell lines (Fig. 2D), with the signal presenting a patterned distribution in the nucleoplasm and a remarkable association with the nuclear lamina during interphase.
Drosphila Piwi shuttles to the nucleous thanks to a N-terminal nuclear localization signal (NLS) that is exposed upon piRNA binding [15]. Interestingly, we identified a predicted NLS in the exon 2 of PIWIL1 (aa 4 to 14), comprising a stretch of basic residues (arginine and alanine). To determine its functionality as a bona fide NLS in Caco-2 cells, we fused this sequence motif to mRuby2. Consistent with the observed predominantly nuclear expression of PIWIL1 in these cells (Fig. 2C), NLS-tagged mRuby2 was mostly expressed in the nucleus (Fig. 2E).
PIWIL1 localizes to the centrosome in Caco-2 cells during mitosis.
While PIWIL1 was mostly localized in the nucleoplasm or in the nuclear lamina in interphasic cells, we consistently observed its expression in the centrosome of mitotic cells (see, for example, Fig. 3). With the purpose of better elucidating the subcellular distribution of PIWIL1 during the cell cycle, we synchronized Caco-2 cells and confirmed PIWIL1 is dynamically redistributed during mitosis (Fig. 3A). Confirming previous observations, synchronized Caco-2 cells co-stained for beta tubulin showed PIWIL1 relocalization to the MTOC during mitosis (Fig. 3B).
Co-localization assays of PIWIL1 and gamma tubulin, a known centrosomal marker, confirmed PIWIL1 as an integral component of the MTOC or the centrosome (Fig. 3C). 3D Rendering of composite images are depicted in Fig. 3D, where PIWIL1 and gamma tubulin appear to be arranged in a manner reminiscent of two interlocking rings, fitting together seamlessly.
PIWIL1 silencing causes G2/M arrest and aberrant mitosis.
To understand the functional implications of PIWIL1 localization to centrosomes in dividing CRC cells, we performed siRNA-mediated PIWIL1 knock down (KD). Levels of PIWIL1 protein and mRNA decreased by 70% as assessed by fluorescence microscopy and RT-qPCR, respectively (Fig. 4A). Surprisingly, KD cells showed a significantly higher frequency of mitotic defects (vs control siRNA-transfected cells), comprising multiple centromeres, anormal mitotic spindles or misaligned metaphasic chromosomes (Fig. 4B). This was followed by cell cycle arrest at G2/M, as measured by flow cytometry using propidium iodide DNA staining (Fig. 4C). These cell division defects were consistent with KD cells having a lower proliferation rate and / or migratory capacity, as measured by wound healing assays (Fig. 4D). In sum, despite Caco-2 cells are transformed and polyploid, they seem to rely on PIWIL1 to regulate mitotic fidelity, potentially avoiding mitotic catastrophe.
PIWIL1 expression is restricted to stem cell niches in normal human colon tissue and is lost upon differentiation
Next, we asked whether PIWIL1 expression in CRC patients and CRC cell lines is due to an aberrant expression of this otherwise germline-specific protein, or whether PIWIL1 is expressed in a subset of human colon cells under physiological conditions. Surprisingly, immunohistochemistry and immunofluorescence microscopy of normal human colon tissue showed PIWIL1 staining restricted to cells at the bottom of each intestinal crypt, where stem cells are known to be located [16] (Fig. 5A). In contrast, colon cancer tissue showed pervasive PIWIL1 staining, with virtually all cells staining positive. However, unlike CRC cell lines, the signal was predominantly cytoplasmic in both healthy and cancer tissues.
Intestinal crypts, which form the base of the villi, are the basic structural and functional units of intestinal epithelium. At the bottom of each crypt, a small number of about eight stem cells [16] are constantly dividing to give place to transit-amplifying progenitor cells. These progenitor cells divide as they migrate upwards and differentiate into two distinct lineages: enterocytes (absorptive cells) and secretory cells.
Having observed that, in normal colon tissue, PIWIL1 expression is restricted to the stem cell population residing at the base of intestinal crypts, we asked whether PIWIL1 expression is downregulated during progenitor cell differentiation into enterocytes. To study this, we followed a well-stablished protocol [17] where Caco-2 cells are differentiated to enterocytes by means of a transwell assay. Surprisingly, unlike 2D cultures of undifferentiated cells (Fig. 2), we were not able to find PIWIL1 expression when Caco-2 cells were differentiated to enterocytes (Fig. 5B). To validate the differentiation protocol, we analyzed the subcellular localization of centrin-2, a structural component of the centrosome, that migrates to the apical level of the cells upon differentiation (Fig. 5B). Thus, loss of PIWIL1 during Caco-2 differentiation resembles the situation in normal colon epithelia, where only undifferentiated cells residing at the base of intestinal crypts express this protein to detectable levels.
To study whether PIWI proteins are required for the maintenance of stemness and self-renewal in non-transformed cells, we analyzed RNA-seq data from GEO: GSE69626. In this study, human fibroblasts were de-differentiated into induced pluripotent stem cells (iPSCs) using six different reprogramming methods [18]. Surprisingly, while PIWIL1 was not involved in this process, PIWIL2 expression was induced in all reprogrammed cells (Fig. 5C). Differentiated fibroblasts expressed FAP but lacked PIWIL2 and the stem cell marker NANOG. Conversely, de-differentiated cells expressed PIWIL2 and NANOG, while silencing FAP.
Albeit involving a different PIWI protein to the one studied herein, results above strengthen the link between PIWI protein expression and stem cell biology. This agrees with PIWIL1 expression in stem cells from healthy colon tissue. In addition, these results potentially explain PIWIL1 overexpression in CRC based on the potential stem cell origin of colorectal cancer [19].