Heterogenous nuclear ribonucleoprotein D-like controls endothelial cell functions
-
Sandra Fischer
, Rolf Backofen
, Kathi Zarnack
and Julia E. Weigand
Abstract
HnRNPs are ubiquitously expressed RNA-binding proteins, tightly controlling posttranscriptional gene regulation. Consequently, hnRNP networks are essential for cellular homeostasis and their dysregulation is associated with cancer and other diseases. However, the physiological function of hnRNPs in non-cancerous cell systems are poorly understood. We analyzed the importance of HNRNPDL in endothelial cell functions. Knockdown of HNRNPDL led to impaired proliferation, migration and sprouting of spheroids. Transcriptome analysis identified cyclin D1 (CCND1) and tropomyosin 4 (TPM4) as targets of HNRNPDL, reflecting the phenotypic changes after knockdown. Our findings underline the importance of HNRNPDL for the homeostasis of physiological processes in endothelial cells.
1 Introduction
Endothelial cells form the inner lining of blood vessels and orchestrate angiogenesis (the formation of new blood vessels from already existing vasculature) in response to external stimuli, such as hypoxia. Angiogenesis is tightly controlled by an interplay of pro- and anti-angiogenic factors and often deregulated in pathological processes, like cancer, where aberrant neovascularization contributes to tumor growth and metastasis (Lugano et al. 2020). Posttranscriptional control is integral to the regulation of angiogenesis, however, so far, only very few RNA-binding proteins (RBPs) are suggested to be involved in angiogenic signaling (Caporali and Emanueli 2011; Chang and Hla 2011; Yu and Wang 2018). RBPs regulate gene expression by controlling alternative splicing, translation efficiency and mRNA stability. Thus, they affect vascular development by controlling the expression of angiogenic factors, such as growth factors, cytokines and cell cycle regulators. The members of the family of heterogeneous nuclear ribonucleoproteins (hnRNPs) are essential regulators in posttranscriptional gene control and often dysregulated in cancer (Sudhakaran and Doseff 2023). However, the function of hnRNPs in angiogenic processes are unclear. Detailed knowledge about the regulation of angiogenesis is essential to be able to inhibit pathological aberrations, such as tumor angiogenesis, and to modulate abnormal vascularization.
In our study, we focus on the ubiquitously expressed HNRNPDL (heterogeneous nuclear ribonucleoprotein D-like), which is implicated in diverse disease settings. HNRNPDL is upregulated in tumor samples of prostate cancer, colon cancer, hepatocellular carcinoma and myeloid leukemia patients (Liu et al. 2007; Wu et al. 2008; Zhang et al. 2018; Zhou et al. 2014). HNRNPDL overexpression transforms B cells and induces leukemia in mice (Ji et al. 2020). Further, dysregulation of HNRNPDL was identified as risk factor for the progression of endometrial cancer (Wang et al. 2019). HNRNPDL belongs to the prion-like RNA-processing proteins, which can aggregate in inclusions implicated in many neurodegenerative diseases (Navarro et al. 2015) and was recently identified as potential biomarker and drug target in depression (Le-Niculescu et al. 2021). Moreover, heterozygous mutations in HNRNPDL cause the muscle-wasting disease limb girdle muscular dystrophy (LGMD) (Berardo et al. 2019; Sun et al. 2019; Vieira et al. 2014). D378H/N mutations occurring in LGMD cause deregulated HNRNPDL phase separation and protein solubility (Batlle et al. 2020; Garcia-Pardo et al. 2023). HNRNPDL was also found to be associated with growth retardation and hypotonia (Hu et al. 2017). However, the cellular role of HNRNPDL in diseases like LGMD, various cancer types and especially in healthy tissues and non-pathological processes is unknown.
HNRNPDL is a paralog of HNRNPD (also AUF1, AU-rich element binding factor 1). HNRNPD is known to regulate the stability and translation efficiency of its mRNA targets by binding to AU-rich elements (AREs) in their 3′-UTR (Yoon et al. 2014, for reviews, see García-Mauriño et al. 2017; White et al. 2017). AREs are abundant cis-regulatory elements tightly controlling mRNA stability. They are found in >50 % of human genes (Bakheet et al. 2018) and are particularly abundant in short-lived mRNAs, encoding growth factors, cytokines and cell cycle regulators. As these factors orchestrate angiogenic processes, ARE-dependent mRNA destabilization will affect angiogenesis. Being recognized by >20 different RBPs, AREs act as interaction hubs in the 3′-UTR of mRNAs, integrating both positive and negative signals to dictate mRNA stability (Plass et al. 2017). The ratio between stabilizing and destabilizing trans-acting factors ultimately determines mRNA fate.
Because paralogs often target similar mRNAs (Smith et al. 2013; Spellman et al. 2007), we hypothesized that HNRNPDL, like its paralog HNRNPD, might affect ARE-mediated mRNA decay.
HNRNPDL protein levels are modulated by alternative splicing (Kemmerer et al. 2018). In previous studies, we could show that HNRNPDL regulates its own expression in a negative feedback loop by binding to its own pre-mRNA in order to enhance the inclusion of the alternative exon 8 (Kemmerer et al. 2018). Exon 8 inclusion initiates decay of the HNRNPDL mRNA via nonsense-mediated mRNA decay (NMD), which subsequently leads to a reduction in protein levels. Further, we could show that chronic hypoxia enhances exon 8 inclusion into the HNRNPDL mRNA in breast cancer cells, reducing HNRNPDL protein levels (Fischer et al. 2020). Also, other cellular stresses, like cisplatin-induced cell death, enhanced exon 8 inclusion in breast cancer cells (Gabriel et al. 2015). Similarly, treatment of human umbilical vein endothelial cells (HUVECs) with the hypoxia mimic CoCl2 led to increased inclusion of HNRNPDL exon 8 (Hang et al. 2009). The regulation of HNRNPDL under stress conditions in endothelial cells and the possible regulation of angiogenic factors by ARE-mediated decay suggest that HNRNPDL is important for HUVEC function. Thus, we explored the cellular function of HNRNPDL in endothelial cells. We identified HNRNPDL to control HUVEC proliferation, migration and sprouting of spheroids. Transcriptome analysis after HNRNPDL knockdown revealed a dual function of HNRNPDL in the regulation of mRNA abundance. In particular, the mRNA of the major cell-cycle regulator cyclin D1 (CCND1) is strongly reduced in the absence of HNRNPDL, while the mRNA of the cytoskeleton protein tropomyosin 4 (TPM4) is increased.
2 Results and discussion
2.1 HNRNPDL regulates endothelial cell functions
In order to investigate whether HNRNPDL affects physiological functions of the endothelium, we analyzed proliferation, migration and sprouting after knockdown of HNRNPDL in HUVECs (Figure 1 and Supplementary Figure S1A). HUVECs were transfected with an siRNA targeting HNRNPDL (si_DLA) or a non-silencing control siRNA (si_ctrl). We achieved a reduction in HNRNPDL protein levels to ∼10 % (Supplementary Figures S1A and S1B). Crystal violet assays were performed to assess changes in proliferation. Knockdown of HNRNPDL decreased HUVEC numbers to 60 % after 48 h and 51 % after 72 h in comparison to the control (Figure 1A). To analyze HUVEC migration, we performed transwell-migration assays. HNRNPDL knockdown dramatically reduced the migration of HUVECs to 16 % (Figure 1B and C). Further, to test how HNRNPDL affects cellular processes important for angiogenesis, we analyzed spheroid sprouting capacity after knockdown of HNRNPDL in 3D cultures. HNRNPDL knockdown reduced spheroid sprouting of HUVECs to 23 % (Figure 1D and E), underlining the importance of HNRNPDL for efficient angiogenesis.

HNRNPDL affects endothelial cell proliferation, migration and sprouting. (A) Cell viability of HUVECs 48 h and 72 h after knockdown of HNRNPDL. n = 3. (B) Quantification of migrating HUVECs after knockdown of HNRNPDL. n = 4. (C) Representative pictures used to quantify the numbers of migrated HUVECs in (B). (D) Quantification of the cumulative sprout length per spheroid in HUVECs after knockdown of HNRNPDL, with or without VEGF-A stimulation (0.05 mg/ml). n = 4. (E) Representative pictures of spheroids and sprouts used to quantify the cumulative sprout length per spheroid in (D). All bar graphs are reported as mean values ± standard deviation. Statistical analysis was done using student’s t-test, two-tailed, paired. (**) p value < 0.01, (*) p value < 0.05, n.s. = not significant.
VEGF-A is a pro-angiogenic factor that stimulates endothelial cell sprouting in blood vessels and promotes spheroid sprouting of HUVECs in cell culture (Yancopoulos et al. 2000). Notably, while VEGF-A stimulation increased sprouting of control cells, it was not able to restore sprouting after HNRNPDL knockdown (Figure 1D and E). Together, the results clearly demonstrate the importance of HNRNPDL for general physiological endothelial cell functions i.e., proliferation, migration and sprouting; processes affecting the formation of new blood vessels.
The observed reduction in HUVEC proliferation after knockdown of HNRNPDL is consistent with its oncogenic function, where HNRNPDL has been shown to promote proliferation in diverse cancer cell types (Ji et al. 2020; Wu et al. 2008; Zhang et al. 2018; Zhou et al. 2014). Thus, the positive effect of HNRNPDL on proliferation seems to be cell type-independent.
In general, RBPs have been suggested to modulate angiogenesis by controlling the expression of cytokines, growth factors and cell cycle regulators. Recently, interleukin enhancer binding factor 3 (ILF-3) and quaking (QKI) were shown to promote angiogenesis by mRNA stabilization. In human coronary artery endothelial cells, ILF-3 induction promoted proliferation, migration and tube formation by stabilizing pro-angiogenic transcripts, such as the mRNA encoding the cytokine IL-8 (CXCL8) (Vrakas et al. 2019). QKI knockdown reduced cell cycle progression and spheroid sprouting in HUVECs, by reducing cell cycle-associated genes. In particular, QKI stabilizes the mRNA encoding CCND1, which promotes G1/S phase transition (Azam et al. 2019). Whether HNRNPDL induces the severe changes in the phenotype of HUVECs by targeting such mRNAs is not known.
2.2 HNRNPDL target mRNAs are involved in cell cycle and motility regulation
In order to identify how HNRNPDL affects endothelial cell function, we performed transcriptomic analysis after HNRNPDL knockdown in HUVECs. To identify the earliest time-point after siRNA transfection when a robust reduction of HNRNPDL was reached, a time-course experiment was performed. This was achieved after 36 h (Figure 2A). Thus, this time point was chosen for RNA sequencing to increase the chance to observe changes mainly in primary targets and minimize secondary effects. Total RNA was extracted from HUVECs transfected with one siRNA targeting HNRNPDL (si_DLA) or a non-silencing control siRNA (si_ctrl), polyA-selected and subjected to deep sequencing (n = 2). Approximately 35 million paired-end reads were obtained for each sample. We detected expression of 13,381 genes with expression being defined as mean transcripts per million [TPM] >1 in either control or HNRNPDL knockdown cells. From these, 3878 genes (29 %) changed significantly in their abundance (adjusted p value < 0.05, Figure 2B and Supplementary Table S1), including 866 >1.5-fold up-regulated and 988 >1.5-fold down-regulated genes.

HNRNPDL regulates target mRNAs involved in cell cycle regulation and cytoskeleton remodeling. (A) Western blot of HNRNPDL and β-actin after siRNA-mediated knockdown for 24, 36 or 48 h. HNRNPDL knockdown for 36 h was chosen as condition for RNA sequencing. n = 2. (B) MA plot showing differentially expressed genes in HNRNPDL knockdown experiments (n = 2). (C) KEGG analysis of the DEGs. (D, E) RT-qPCR quantification of CCND1 and TPM4 after HNRNPDL knockdown in HUVECs with si_DLA (D, n = 4) and si_DLB (E, n = 3). All bar graphs are reported as mean values ± standard deviation. Statistical analysis was done using student’s t-test, two-tailed, paired. (**) p value < 0.01, (*) p value < 0.05.
Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of the differentially expressed genes was used to identify significantly changed pathways in response to HNRNPDL knockdown (Figure 2C and Supplementary Table S2). With an adjusted p-value of 3.37 × 10−8, “cell cycle” was most significantly affected. Interestingly, the mRNA of the major cell cycle regulator cyclin D1 (CCND1) controlling G1/S phase transition, was the most highly reduced mRNA amongst cell cycle genes (Figure 2C, red dot). CCND1 controls cell cycle progression by interaction with cycline-dependent kinases 4 or 6. In cancer, CCND1 is often upregulated and leads to excessive cell proliferation (reviewed in Tchakarska and Sola 2020). Thus, reduction of CCND1 mRNA is in concordance with the observed reduction in proliferation after HNRNPDL knockdown. We used RT-qPCR to verify and quantify the reduced mRNA levels of CCND1 in response to HNRNPDL knockdown. Two different siRNAs (si_DLA and si_DLB) were transfected to ensure that the observed changes are no off-target effects of the siRNA (si_DLA) used for RNA-seq. The knockdown of HNRNPDL with si_DLA as well as si_DLB was confirmed via Western Blot (Supplementary Figure S1A). HNRNPDL protein levels were reduced to ∼10 % for si_DLA and to ∼13 % for si_DLB (Supplementary Figure S1B). RT-qPCR verified the expected reduction in CCND1 mRNA abundance after knockdown of HNRNPDL with both siRNAs (Figure 2D and E).
Further, we confirmed induction of the most abundantly expressed tropomyosin, TPM4, at mRNA as well as protein level by RT-qPCR and Western Blot, respectively (Figure 2D and E, Supplementary Figure S2). Tropomyosins affect cell migration by modulating the dynamics of actin cytoskeleton, which forms along the leading edge of a migrating cell (Khaitlina 2015; Schaks et al. 2019). Interestingly, TPM4 is discussed as biomarker in multiple cancer types, with contrasting results regarding its influence on cell migration and disease prognosis, which might depend on the identity of TPM4 protein isoforms (Jeong et al. 2017; Luo et al. 2022; Sheng and Chen 2020; Yang et al. 2018; Zhao et al. 2019). TPM4 induction in HUVECs is concordant with reduced cell migration after HNRNPDL knockdown, as reported for colon, cervical and breast cancer (Jeong et al. 2017; Luo et al. 2022; Yang et al. 2018).
2.3 Dual regulation by HNRNPD and HNRNPDL paralogs
Paralogous RBPs often affect similar mRNA targets (Rahman et al. 2013; Vuong et al. 2016). Further, paralogs tend to control each other’s expression by cross-regulation. Likewise, HNRNPD and DL control their own as well as each other via alternative splicing of poison exons in their 3′-UTRs (Figure 3A, indicated in red). Cross-regulation of HNRNPD expression by HNRNPDL is exerted by controlled inclusion of exon 9 into the HNRNPD mRNA. Inclusion of exon 9 subsequently triggers degradation of the mRNA by NMD (Kemmerer et al. 2018). Knockdown of HNRNPDL is expected to reduce exon 9 inclusion into the HNRNPD mRNA and thus, to increase HNRNPD protein levels, due to the reduced production of NMD-targeted mRNA isoforms.

Dual regulation of CCND1 and TPM4 by HNRNPD and DL. (A) Exon cluster of HNRNPD and HNRNPDL. (B) RT-qPCR quantification of HNRNPDL, HNRNPD, CCND1, and TPM4 after double knockdown of HNRNPDL and HNRNPD in HUVECs all bar graphs are reported as mean values ± standard deviation. Statistical analysis was done using student’s t-test, two-tailed, paired. (**) p value < 0.01, n.s. = not significant. n = 4–5. (C and D) 12-mer motifs derived by GraphProt (Maticzka et al. 2014) analysis from RBNS data for HNRNPD (C) and HNRNPDL (D). (E and F) Correlation of HNRNPDL, HNRNPD, ZFP36, and ELAVL1.
To analyze if the observed changes in target mRNA levels could be due to altered HNRNPD protein levels, we analyzed exon 9 inclusion levels and the levels of all four HNRNPD protein isoforms (Figure 3A and Supplementary Figure S3). Indeed, knockdown of HNRNPDL led to reduced inclusion of HNRNPD exon 9 in HUVECs (Supplementary Figure S3A). However, Western blot analysis showed that HNRNPD protein levels were not changed after HNRNPDL knockdown (Supplementary Figure S3B). This finding is most likely explained by the extremely low basal expression of exon 9-including mRNA isoforms in HUVECs. Thus, a further reduction in exon 9-containing isoforms does not significantly increase HNRNPD protein levels. In sum, although the cross-regulation of HNRNPD expression by HNRNPDL is functional in HUVECs, HNRNPD is not the driver of the observed changes in CCND1 and TPM4 mRNA abundances.
To examine if HNRNPD protein reduction can affect HNRNPDL target mRNA abundances in HUVECs, we investigated the impact of simultaneous HNRNPD and DL knockdown on the levels of CCND1 and TPM4 mRNAs. HUVECs were transfected with identical amounts of siRNAs: control (si_ctrl), control and HNRNPDL (si_ctrl+si_DLA), and HNRNPDL and HNRNPD (si_DLA+si_D). HNRNPD levels increased with HNNRPDL knockdown in line with the reduced exon 9 inclusion and strongly decreased in the double knockdown (Figure 3B). This change in mRNA abundance of HNRNPD is also reflected in HNRNPD protein levels (Supplementary Figure S4).
HNRNPD has been shown to be a negative regulator of CCND1 in several cell types including cervical carcinoma (HeLa), human osteosarcoma (U2OS), colon carcinoma (HCT116) and adenocarcinoma (LoVo) as well as primary normal human skin fibroblasts HFSN1 (Al-Khalaf et al. 2011; Lal et al. 2004; Tian et al. 2020). This suggests that simultaneous HNRNPD knockdown could potentially counteract the reduction in CCND1 mRNA observed after HNRNPDL knockdown. However, CCND1 mRNA was not affected by the additional HNRNPD reduction in HUVECs (Figure 3B). Thus, CCND1 might either be not targeted by HNRNPD in HUVECs or is more dependent on HNRNPDL status. Notably, regulation of CCND1 levels by HNRNPD were shown to be complex, with varying results found in different cellular systems and at different HNRNPD levels. One reason for this might be that the CCND1 mRNA is targeted by a multitude of stabilizing and destabilizing RBPs as well as lncRNAs and miRNAs, the sum of which will ultimately determine CCND1 mRNA levels (Al-Khalaf et al. 2011; Azam et al. 2019; Cao et al. 2017; Chen et al. 2019, 2008; Ghosh and Adhya 2018; Lal et al. 2004; Liu et al. 2021; Yoneda et al. 2020).
In contrast, not much is known about the posttranscriptional regulation of TPM4. PTB (polypyrimidine tract binding protein) destabilizes the TPM4 mRNA when bound by the lncRNA SFTA1P in cervical cancer cell lines (Luo et al. 2022). ARE-mediated decay of TPM4 has not been investigated so far. Interestingly, TPM4 mRNA induction was significantly increased by double knockdown of HNRNPD and DL compared to the knockdown of HNRNPDL alone (Figure 3B). Thus, the two paralogs can act additively in mRNA reduction. This contrasting behavior of HNRNPD and DL towards different targets is reminiscent of other paralogs such as HNRNPL and LL as well as PTB and nPTB, which can act redundant, additive or even competitive on shared targets (Rahman et al. 2013; Vuong et al. 2016). Here, the outcome in target regulation can be the result of subtle differences in RNA-binding preferences between the paralogs.
To evaluate this further, we analyzed publicly available RNA Bind-n-Seq (RBNS) data (Van Nostrand et al. 2020) to compare the RNA-binding preferences of HNRNPD and HNRNPDL. Using GraphProt (Maticzka et al. 2014), models were trained using default parameters on samples of 100,000 training sequences and tested with a 10-fold cross-validation. The obtained binding motif for HNRNPD showed the expected very strong preference for adenine and uridine residues as typically found in AREs (Figure 3C). The binding preference is highly similar to the one from HNRNPDL (Figure 3D). This is expected, as the two paralogs share highly similar RNA-binding domains (>74 % sequence homology), which is indicative for similar motif preferences (Ray et al. 2013). Direct comparison of the top ten RBNS hexamers identified for HNRNPD and HRNPDL indicates an elevated preference of HNRNPDL for longer A-stretches in this ARE motifs (Supplementary Table S3).
Next, we visualized the top ten RBNS hexamers in the 3′-UTRs of the CCND1 and TPM4 mRNAs as potential binding sites for HNRNPD and DL (Supplementary Figures S5 and S6). A 9 nt long HNRNPDL binding site resulting from overlapping hexamer motifs is present in a 470 nucleotide long AU-rich region of the 3′-UTR of CCND1, that was previously described to increase CCND1 mRNA stability (Deshpande et al. 2009). Thus, HNRNPDL likely is able to interact with this regulatory region, subsequently modulating mRNA abundance. Interestingly, several high scoring HNRNPD hexamers are also present, in this AU-rich region, in line with regulation of CCND1 mRNA levels by HNRNPD observed in other cells types.
The 1656 nt long TPM4 3′-UTR (hg38_knownGene_ENST00000643579.2 Supplementary Figure S6), contains fifteen hexamer motifs of HNRNPDL and two overlapping ones of HNRNPD. These overlapping hexamers can also be recognized by HNRNPDL (Supplementary Figure S6). Thus, the TPM4 3′-UTR contains recognition motifs of both HNRNPD and DL allowing for the additive regulation observed in the knockdown experiments. In sum, both 3′-UTRs contain several of the AU-rich hexamers identified as recognition motifs of HNRNPD and DL by RBNS, allowing for dual regulation.
To get a more detailed insight into the correlation of HNRNPD and DL in relation to other ARE-binding proteins, we first examined publicly available single cell transcriptome data from the Genotype-Tissue Expression (GTEx) Project (GTEx Consortium 2020) to identify a tissue with highly expressed HNRNPDL in healthy cells. Single cell tissue gene expression for HNRNPDL (ENSG00000152795.17) in vascular endothelial cells (Supplementary Figure S7A) is highest in breast mammary tissue. Thus, we used breast tissue data to analyze the relation between the expression of HNRNPDL, HNRNPD and two other ARE-binding RBPs, ZFP36 (zinc finger protein 36, also TTP) and ELAVL1 (ELAV like RNA-binding protein 1, also HuR) (Figure 3E, Supplementary S7B and S7C). As expected, the expression of HNRNPD and DL is highly positively correlated (Supplementary Figures S7C and S7E), which likely reflects the cross-regulation by AS-NMD. Further, HNRNPDL levels are also positively correlated with ZFP36 or ELAVL1, albeit two-fold lower than with HNRNPD. Further, we compared expression levels in breast cancer tissue to see if RBP networks are disturbed under pathological conditions (https://www.cancer.gov/tcga) (Figure 3F and Supplementary Figures S7D and S7E). In general, the positive correlation between the different RBPs decreases in tumor tissue, suggestive of pathological alterations in RBP levels (Figure 3E and F).
Together, the data suggest that AREs and ARE-binding RBPs, such as HNRNPDL, affect angiogenic processes by modulating mRNA abundances. The comparison of healthy and tumor tissue demonstrates the changing RBP landscape, which in turn will affect mRNA levels in a cell. However, how differential target regulation is achieved and differs depending on the cell type is still unclear and warrants further investigation.
3 Significance
The results from physiological tests and transcriptomic analysis demonstrate a major role of HNRNPDL in the endothelium. It affects cell proliferation and migration, by regulating the mRNA levels of cell cycle and cell motility factors. RBPs and RNA-protein interactions have the potential to serve as novel drug targets for therapeutic intervention in cancer and other diseases involving aberrant angiogenesis. Future work needs to address the interplay between HNRNPDL and other ARE-binding proteins and their role in physiological cell systems.
4 Materials and methods
4.1 Cell culture and transfection
HUVECs (Lonza, CC-2519) were cultured in T75 flasks in EBM medium (Lonza) supplemented with 10 % FCS (Thermo Fisher Scientific) and EGM single quots [hEGF, Hydrocortison, GA-1000, bovine brain extract (Lonza)]. HUVECs were split once a week and only passages 2 and 3 were used for experiments. For siRNA transfection, 3.5 × 105 cells were seeded in 60 mm cell culture dishes. 24 h later, transfection was performed using Lipofectamine RNAiMAX according to the manufacturer’s protocol with varying amounts of siRNA (60 pmol siRNA for single knockdown, 120 pmol for double knockdown) in OptiMEM medium. After 4 h the transfection mixes were removed, and fresh medium was added.
Target | Sequence | |
---|---|---|
si_ctrl | Non-silencing | 5′-UUCUCCGAACGUGUCACGU[dT][dT]-3′ |
si_DLA | HNRNPDL | 5′-GGGUAUAACUAUGGGAACU[dT][dT]-3′ |
si_DLB | HNRNPDL | 5′-AAGTGTGAAATCAAAGTTGCA[dT][dT]-3′ |
si_D | HNRNPD | 5′-GAAGGUGAUUGAUCCUAAA[dT][dT]-3′ |
4.2 RNA isolation
Total RNA from HUVECs was isolated using the miRNeasy Mini kit (Qiagen), including the optional on-column DNA digest with the RNase-Free DNase Set (Qiagen). After isolation, 500 ng RNA were quality checked on a 1 % agarose gel.
4.3 RNA sequencing
For the HNRNPDL knockdown experiments (n = 2), total RNA from HUVECs transfected either with a non-silencing control siRNA (si_ctrl) or one siRNA targeting HNRNPDL (si_DLA) was used to generate polyA-enriched strand-specific cDNA libraries. Libraries were sequenced on a HiSeq 2000 (Illumina), obtaining 76 nt paired-end reads. Approximately 35 million reads were obtained for each library.
4.4 RNA-seq data analyses
Reads were mapped to the human genome (GRCh38/hg38 assembly) using the splice-aware aligner STAR version 2.5.4b (Dobin et al. 2013). The htseq-count script from the HTSeq python package version 0.6.1p1 (Anders et al. 2015) was used to count reads within genes annotated in GENCODE version 32. Differential expression analysis between normal and HNRNPDL knockdown conditions was performed using the R/Bioconductor package DESeq2 (Love et al. 2014) using the method apeglm for effect size shrinkage (Zhu et al. 2019). Here, differentially expressed genes were defined based on an adjusted p value < 0.05 and a shrunken absolute fold change >1.5. This identified 1854 significantly differentially regulated genes, including 866 upregulated and 988 downregulated genes. For all analyses, we considered only expressed genes with a mean expression level of >1 transcript per million (TPM) in at least one of the two conditions and at least 10.23 mean normalized read counts (baseMean).
4.5 KEGG analysis
The KEGG enrichment analysis was performed with clusterProfiler (v. 3.16.0) (Yu et al. 2012) for the genes that were significantly upregulated (n = 1924) or downregulated (n = 1954). 64 KEGG pathways were significantly enriched (adjusted p value < 0.05, Supplementary Table S2). Selected enriched pathways are shown in Figure 2C.
4.6 RBNS data analysis
We used GraphProt (Maticzka et al. 2014), which was developed with CLIP-seq and RNAcompete data in mind, to find motif enrichments from RBNS data for HNRNPD and HNRNPDL, respectively (ENCODE project; experiment IDs: ENCSR175OMA, ENCSR055HDN (Van Nostrand et al. 2020)). We used the negative controls (ENCSR141NLX, ENCSR394HHV, ENCSR599EMY (Van Nostrand et al. 2020)) to train a classification model and used randomly 100,000 training sequences for the training phase. We validated the models with a 10-fold cross-validation.
4.7 RT-qPCR analyses
RT-qPCR quantification was performed as in Weigand et al. (2012). Oligonucleotide sequences are listed in Supplementary Table S4. All PCR products were verified by sequencing.
4.8 Western blotting
HUVECs were lysed in lysis buffer [137 mM NaCl, 10 % glycerol, 20 mM Tris-HCl pH 8.0, 2 mM EDTA pH 8.0, 1 % Igepal, 5 μl protease inhibitor cocktail (Sigma-Aldrich, St. Louis, USA)] for 20 min on ice. The samples were centrifuged (15 min at 17,000 g, 4 °C) and cell debris was removed. The protein content of the samples was determined in three technical replicates according to the Bradford method. 10 μg protein were loaded onto 10 % SDS polyacrylamide gels. Total lane protein (loading control) was visualized using 2,2,2-trichloroethanol (Ladner et al. 2004). Subsequently, proteins were blotted onto PVDF membranes (Bio-Rad, Hercules, USA) and membranes were blocked using 2 % skim milk. Primary antibodies are listed in Supplementary Table S5 Horseradish peroxidase-conjugated anti-mouse or anti-rabbit IgG (Jackson ImmunoResearch, West Grove, USA) were used as secondary antibodies. Blots were developed with the ECL system (Bio-Rad) or Amersham ECL select (GE Healthcare, Chicago, USA) for weaker signals. Images were detected using the ChemiDoc Imaging System (Bio-Rad). The Western Blots were quantified using Image Lab v 6.1.0 build 7 (Bio-Rad).
4.9 Cell viability
HUVECs were transfected as described above. 48 or 72 h after transfection, HUVECs were fixed with 0.5 % formaldehyde in PBS and stained with 0.5 % crystal violet in PBS. After three washing steps with PBS the cells were incubated with 33 % acetic acid. Samples were transferred to a 96-well plate. Absorption at 620 nm was measured in a TECAN infinite M 200 Pro plate reader (Tecan, Männedorf, Switzerland). Absorption was normalized to untreated HUVEC control cells.
4.10 HUVEC migration assay
24 h after transfection with si_ctrl or si_DLA, HUVECs were serum-starved [EBM medium without FCS (Lonza Group, Basel, Switzerland)]. Again 24 h later, HUVECs were washed with 1× PBS and detached using Trypsin-EDTA [0.05 % trypsin, 0.02 % EDTA in 1× PBS (Thermo Fisher Scientific, Waltham, USA)]. HUVECs were resuspended in starvation medium (EBM medium without FCS). 5 × 104 cells in 100 μl starvation medium were seeded per insert in a 24-well transwell chamber [Thincert cell culture inserts, pore diameter 8 μm, translucent PET membrane (Greiner Bio-One, Kremsmünster, Austria)]. Transwell inserts were placed into a lower well containing 700 μl EBM medium with supplements or starvation medium as a negative control. After 5 h (37 °C, 5 % CO2), cells were removed from the inner side of the insert with a cotton swab. Migrated cells were washed with 1× PBS and fixed with methanol. Migrated cells were stained using crystal violet (0.5 % in 1× PBS) and the membranes were mounted using Pertex (Histolab, Gothenburg, Norway). No cells were detected in the negative control. Migrated cells in six fields per well from random sites of the transwell insert membrane were counted using Fiji software (version 1.51f).
4.11 HUVEC spheroid sprouting assay
24 h after transfection, HUVECs were detached using Trypsin-EDTA (0.05 % trypsin, 0.02 % EDTA in 1× PBS). 500 HUVECs were seeded per 50 μl drop as hanging drop culture in EBM medium supplemented with 20 % EBM-methylcellulose medium [1.2 % (w/v) methylcellulose (Carl Roth, Karlsruhe, Germany) in EBM medium without supplements]. Approximately 100 drops were created for each condition (si_ctrl or si_DLA). 24 h later, spheroids were transferred to a collagen matrix. Collagen (Sigma-Aldrich) was mixed with 10 % 10× M199 medium (Sigma-Aldrich). The collagen mix was prepared for all samples, so that the pH is the same for every condition. Spheroids were harvested in 1× PBS and centrifuged for 3 min with 500 g. The supernatant was removed. 2.6 ml methylcellulose-FCS (20 % FCS, 80 % EBM-methylcellulose medium) was added to the spheroids. The samples (si_ctrl or si_DLA) were split between two reaction tubes. Spheroids in one of the two reaction tubes were stimulated with 0.05 mg/ml VEGF-A protein (VEGF165, Peprotech). The collagen medium mix was then neutralized using 0.2 M NaOH. 1.3 ml collagen mix was added to each spheroid sample. Samples were transferred immediately into the inner wells of a 24-well plate (4 wells per sample) without producing air bubbles and incubated for 24 h at 37 °C, 5 % CO2. Outer wells of the 24-well plate were filled with 1× PBS beforehand to prevent draining of the spheroids. 24 h later, spheroids and their sprouts were photographed using a Zeiss (Oberkochen, Germany) Axiovert 200 microscope (10× objective). The cumulative sprout length in µm of 10 spheroids per condition and biological replicate was measured using Fiji software (version 1.51f).
4.12 Statistical analyses
All bar graphs are reported as mean values ± standard deviation. Statistical analysis was done using student’s t-test, two-tailed, paired.
4.13 Bioinformatic analysis of HNRNPDL expression and correlation
We used public RNA expression data from breast tissue (GTEx, (GTEx Consortium 2020)) and breast cancer tissue (data generated by the TCGA Research Network: https://www.cancer.gov/tcga) to compare physiological and pathological expression of HNRNPDL to HNRNPD, ZPF36, and ELAVL1.
Data was obtained using the recount3 v 1.8.0 (Wilks et al. 2021) package from Bioconductor using the create_rse function and then the compute_read_counts function. The breast tissue data was selected with the keywords file_source: “gtex”, project_type: “data_sources” and project: “BREAST”. The breast tumor data was selected with project: “BRCA”. From the breast cancer samples only samples from female donors were used, only the sampleType: “Primary Tumor” was used and only samples with one sample per donor were used. If more than one tumor sample exists from the same donor, all are removed.
Both the breast tissue data and the breast cancer data were processed following the following steps to obtain comparable RPKM measurements: (i) Genes with low expression were filtered with the filterByExpr function from edgeR Bioconductor package v 3.40.2 (McCarthy et al. 2012) with default filter settings. (ii) For breast tissue data, batch effect correction was performed on the batches provided by GTEx (smnabtcht). For batch correction the ComBat_seq algorithm from the sva Bioconductor package v 3.46.0 (Leek et al. 2023) was used. (iii) TMM values were calculated from the read count data by dividing the read counts by a TMM normalization factor, which was calculated with calcNormFactors from edgeR. (iv) Then RPKM values were calculated with edgeR rpkm function setting log = TRUE.
Expression of HNRNPDL to HNRNPD, ZPF36, and ELAVL1 is shown in box-and-whisker plots. Correlations are calculated as Pearson correlations. Dotplots are made with the corrplot package (v 0.92). Pearson’s R is given as color scale and the areas of circles show the absolute value of corresponding correlation coefficients. For correlations with a p-value > 0.001 no dot is shown.
4.14 Accession numbers
The RNA-seq data of the HNRNPDL knockdown experiments have been deposited with the Gene Expression Omnibus database (GEO; www.ncbi.nlm.nih.gov/geo/) and are available under the accession number GSE169378.
Funding source: Dr. Hans Messer Stiftung
Funding source: Deutsche Forschungsgemeinschaft
Award Identifier / Grant number: GRK2344/322977937
Award Identifier / Grant number: SFB902/B13
Award Identifier / Grant number: SFB902/B14
Award Identifier / Grant number: TRR267/A01
Funding source: Dr. Ing. Wilhelm und Maria Kirmser-Stiftung
Acknowledgments
We thank Professor Dr. Beatrix Suess for the helpful discussions and Anna Theresa Gimbel and Britta Kluge for excellent assistance.
-
Research ethics: Not applicable.
-
Author contributions: The authors have accepted responsibility for the entire content of this manuscript and approved its submission. S.F., C.L., K.Z. and J.E.W. conceptualized research experiments and goals. S.F. and C.L. planned and performed the majority of the experiments. K.K. prepared sequencing samples and performed Western Blot in Figure 2A. A.S., M.K. and K.Z. performed bioinformatic analysis of RNA-seq data and tissue expression of RBPs. F.H., D.M. and R.B. performed bioinformatic analysis of RBNS data. S.F., C.L. and J.E.W. wrote the manuscript with input from all authors.
-
Competing interests: The authors state no conflict of interest.
-
Research funding: This work was supported by the Deutsche Forschungsgemeinschaft [SFB902/B14 to J.E.W.; SFB902/B13 and TRR267/A01 to Z.K.; GRK2344/322977937 to F.H.], the Dr. Hans Messer Stiftung [to J.E.W.] and the Dr. Ing. Wilhelm und Maria Kirmser-Stiftung [to S.F.].
-
Data availability: The raw data can be obtained on request from the corresponding author. The RNA-seq data of the HNRNPDL knockdown experiments have been deposited with the Gene Expression Omnibus database (GEO; www.ncbi.nlm.nih.gov/geo/) and are available under the accession number GSE169378.
References
Al-Khalaf, H.H., Colak, D., Al-Saif, M., Al-Bakheet, A., Hendrayani, S.-F., Al-Yousef, N., Kaya, N., Khabar, K.S., and Aboussekhra, A. (2011). p16INK4A positively regulates cyclin D1 and E2F1 through negative control of AUF1. PLOS ONE 6: e21111, https://doi.org/10.1371/journal.pone.0021111.Search in Google Scholar PubMed PubMed Central
\Anders, S., Pyl, P.T., and Huber, W. (2015). HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31: 166–169, https://doi.org/10.1093/bioinformatics/btu638.Search in Google Scholar PubMed PubMed Central
Azam, S.H., Porrello, A., Harrison, E.B., Leslie, P.L., Liu, X., Waugh, T.A., Belanger, A., Mangala, L.S., Lopez-Berestein, G., Wilson, H.L., et al.. (2019). Quaking orchestrates a post-transcriptional regulatory network of endothelial cell cycle progression critical to angiogenesis and metastasis. Oncogene 38: 5191–5210, https://doi.org/10.1038/s41388-019-0786-6.Search in Google Scholar PubMed PubMed Central
Bakheet, T., Hitti, E., Al-Saif, M., Moghrabi, W.N., and Khabar, K.S.A. (2018). The AU-rich element landscape across human transcriptome reveals a large proportion in introns and regulation by ELAVL1/HuR. Biochim. Biophys. Acta BBA - Gene Regul. Mech. 1861: 167–177, https://doi.org/10.1016/j.bbagrm.2017.12.006.Search in Google Scholar PubMed
Batlle, C., Yang, P., Coughlin, M., Messing, J., Pesarrodona, M., Szulc, E., Salvatella, X., Kim, H.J., Taylor, J.P., and Ventura, S. (2020). hnRNPDL phase separation is regulated by alternative splicing and disease-causing mutations accelerate its aggregation. Cell Rep 30: 1117–1128.e5, https://doi.org/10.1016/j.celrep.2019.12.080.Search in Google Scholar PubMed PubMed Central
Berardo, A., Lornage, X., Johari, M., Evangelista, T., Cejas, C., Barroso, F., Dubrovsky, A., Bui, M.T., Brochier, G., Saccoliti, M., et al.. (2019). HNRNPDL-related muscular dystrophy: expanding the clinical, morphological and MRI phenotypes. J. Neurol. 266: 2524–2534, https://doi.org/10.1007/s00415-019-09437-3.Search in Google Scholar PubMed
Cao, L., Zhang, P., Li, J., and Wu, M. (2017). LAST, a c-Myc-inducible long noncoding RNA, cooperates with CNBP to promote CCND1 mRNA stability in human cells, (I. Ulitsky, ed.). eLife 6: e30433.10.7554/eLife.30433Search in Google Scholar PubMed PubMed Central
Caporali, A. and Emanueli, C. (2011). MicroRNA regulation in angiogenesis. Vascul. Pharmacol. 55: 79–86, https://doi.org/10.1016/j.vph.2011.06.006.Search in Google Scholar PubMed
Chang, S.-H. and Hla, T. (2011). Gene regulation by RNA binding proteins and microRNAs in angiogenesis. Trends Mol. Med. 17: 650–658, https://doi.org/10.1016/j.molmed.2011.06.008.Search in Google Scholar PubMed PubMed Central
Chen, G., Hu, M., Qu, X., Wang, K., and Qu, Y. (2019). MicroRNA-584 directly targets CCND1 and inhibits cell proliferation and invasion in pancreatic cancer. Mol. Med. Rep 19: 719–726, https://doi.org/10.3892/mmr.2018.9651.Search in Google Scholar PubMed
Chen, R.W., Bemis, L.T., Amato, C.M., Myint, H., Tran, H., Birks, D.K., Eckhardt, S.G., and Robinson, W.A. (2008). Truncation in CCND1 mRNA alters miR-16-1 regulation in mantle cell lymphoma. Blood 112: 822–829, https://doi.org/10.1182/blood-2008-03-142182.Search in Google Scholar PubMed PubMed Central
Deshpande, A., Pastore, A., Deshpande, Aniruddha, J., Zimmermann, Y., Hutter, G., Weinkauf, M., Buske, C., Hiddemann, W., and Dreyling, M. (2009). 3′UTR mediated regulation of the cyclin D1 proto-oncogene. Cell Cycle 8: 3592–3600, https://doi.org/10.4161/cc.8.21.9993.Search in Google Scholar PubMed
Dobin, A., Davis, C.A., Schlesinger, F., Drenkow, J., Zaleski, C., Jha, S., Batut, P., Chaisson, M., and Gingeras, T.R. (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29: 15–21, https://doi.org/10.1093/bioinformatics/bts635.Search in Google Scholar PubMed PubMed Central
Fischer, S., Liddo, A.D., Taylor, K., Gerhardus, J.S., Sobczak, K., Zarnack, K., and Weigand, J.E. (2020). Muscleblind-like 2 controls the hypoxia response of cancer cells. RNA 26: 648–663, https://doi.org/10.1261/rna.073353.119.Search in Google Scholar PubMed PubMed Central
Gabriel, M., Delforge, Y., Deward, A., Habraken, Y., Hennuy, B., Piette, J., Klinck, R., Chabot, B., Colige, A., and Lambert, C. (2015). Role of the splicing factor SRSF4 in cisplatin-induced modifications of pre-mRNA splicing and apoptosis. BMC Cancer 15: 227, https://doi.org/10.1186/s12885-015-1259-0.Search in Google Scholar PubMed PubMed Central
García-Mauriño, S.M., Rivero-Rodríguez, F., Velázquez-Cruz, A., Hernández-Vellisca, M., Díaz-Quintana, A., De la Rosa, M.A., and Díaz-Moreno, I. (2017). RNA binding protein regulation and cross-talk in the control of AU-rich mRNA fate. Front. Mol. Biosci. 4: 71, https://doi.org/10.3389/fmolb.2017.00071.Search in Google Scholar PubMed PubMed Central
Garcia-Pardo, J., Bartolomé-Nafría, A., Chaves-Sanjuan, A., Gil-Garcia, M., Visentin, C., Bolognesi, M., Ricagno, S., and Ventura, S. (2023). Cryo-EM structure of hnRNPDL-2 fibrils, a functional amyloid associated with limb-girdle muscular dystrophy D3. Nat. Commun. 14: 239, https://doi.org/10.1038/s41467-023-35854-0.Search in Google Scholar PubMed PubMed Central
Ghosh, U. and Adhya, S. (2018). Posttranscriptional regulation of cyclin D1 by ARE-binding proteins AUF1 and HuR in cycling myoblasts. J. Biosci. 43: 685–691, https://doi.org/10.1007/s12038-018-9788-8.Search in Google Scholar
GTEx Consortium. (2020). The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369: 1318–1330, https://doi.org/10.1126/science.aaz1776.Search in Google Scholar PubMed PubMed Central
Hang, X., Li, P., Li, Z., Qu, W., Yu, Y., Li, H., Shen, Z., Zheng, H., Gao, Y., Wu, Y., et al.. (2009). Transcription and splicing regulation in human umbilical vein endothelial cells under hypoxic stress conditions by exon array. BMC Genom. 10: 126, https://doi.org/10.1186/1471-2164-10-126.Search in Google Scholar PubMed PubMed Central
Hu, X., Chen, X., Wu, B., Soler, I.M., Chen, S., and Shen, Y. (2017). Further defining the critical genes for the 4q21 microdeletion disorder. Am. J. Med. Genet. A. 173: 120–125, https://doi.org/10.1002/ajmg.a.37965.Search in Google Scholar PubMed
Jeong, S., Lim, S., Schevzov, G., Gunning, P.W., and Helfman, D.M. (2017). Loss of Tpm4.1 leads to disruption of cell-cell adhesions and invasive behavior in breast epithelial cells via increased Rac1 signaling. Oncotarget 8: 33544–33559, https://doi.org/10.18632/oncotarget.16825.Search in Google Scholar PubMed PubMed Central
Ji, D., Zhang, P., Ma, W., Fei, Y., Xue, W., Wang, Y., Zhang, X., Zhou, H., and Zhao, Y. (2020). Oncogenic heterogeneous nuclear ribonucleoprotein D-like modulates the growth and imatinib response of human chronic myeloid leukemia CD34+ cells via pre-B-cell leukemia homeobox 1. Oncogene 39: 443–453, https://doi.org/10.1038/s41388-019-0998-9.Search in Google Scholar PubMed
Kemmerer, K., Fischer, S., and Weigand, J.E. (2018). Auto- and cross-regulation of the hnRNPs D and DL. RNA 24: 324–331, https://doi.org/10.1261/rna.063420.117.Search in Google Scholar PubMed PubMed Central
Khaitlina, S.Yu. (2015). Tropomyosin as a regulator of actin dynamics (Chapter Seven). In: Jeon, K.W. (Ed.). International review of cell and molecular biology. Academic Press, Cambridge, Massachusetts, USA, pp. 255–291.10.1016/bs.ircmb.2015.06.002Search in Google Scholar PubMed
Ladner, C.L., Yang, J., Turner, R.J., and Edwards, R.A. (2004). Visible fluorescent detection of proteins in polyacrylamide gels without staining. Anal. Biochem. 326: 13–20, https://doi.org/10.1016/j.ab.2003.10.047.Search in Google Scholar PubMed
Lal, A., Mazan-Mamczarz, K., Kawai, T., Yang, X., Martindale, J.L., and Gorospe, M. (2004). Concurrent versus individual binding of HuR and AUF1 to common labile target mRNAs. EMBO J. 23: 3092–3102, https://doi.org/10.1038/sj.emboj.7600305.Search in Google Scholar PubMed PubMed Central
Leek, J.T., Johnson, W.E., Parker, H.S., Fertig, E.J., Jaffre, A.E., Zhang, Y., Storey, J.D., and Torres, L.C. (2023). sva: Surrogate variable analysis. R package version 3.46.0.Search in Google Scholar
Le-Niculescu, H., Roseberry, K., Gill, S.S., Levey, D.F., Phalen, P.L., Mullen, J., Williams, A., Bhairo, S., Voegtline, T., Davis, H., et al.. (2021). Precision medicine for mood disorders: objective assessment, risk prediction, pharmacogenomics, and repurposed drugs. Mol. Psychiatry: 1–29, https://doi.org/10.1038/s41380-021-01061-w.Search in Google Scholar PubMed PubMed Central
Liu, Y., Zhang, A., Bao, P.-P., Lin, L., Wang, Y., Wu, H., Shu, X.-O., Liu, A., and Cai, Q. (2021). MicroRNA-374b inhibits breast cancer progression through regulating CCND1 and TGFA genes. Carcinogenesis 42: 528–536, https://doi.org/10.1093/carcin/bgab005.Search in Google Scholar PubMed PubMed Central
Liu, Y., Zhu, X., Zhu, J., Liao, S., Tang, Q., Liu, K., Guan, X., Zhang, J., and Feng, Z. (2007). Identification of differential expression of genes in hepatocellular carcinoma by suppression subtractive hybridization combined cDNA microarray. Oncol. Rep. 18: 943–951, https://doi.org/10.3892/or.18.4.943.Search in Google Scholar
Love, M.I., Huber, W., and Anders, S. (2014). Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15: 550, https://doi.org/10.1186/s13059-014-0550-8.Search in Google Scholar PubMed PubMed Central
Lugano, R., Ramachandran, M., and Dimberg, A. (2020). Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. CMLS 77: 1745–1770, https://doi.org/10.1007/s00018-019-03351-7.Search in Google Scholar PubMed PubMed Central
Luo, A., Lan, X., Qiu, Q., Zhou, Q., Li, J., Wu, M., Liu, P., Zhang, H., Lu, B., Lu, Y., et al.. (2022). LncRNA SFTA1P promotes cervical cancer progression by interaction with PTBP1 to facilitate TPM4 mRNA degradation. Cell Death Dis. 13: 936, https://doi.org/10.1038/s41419-022-05359-7.Search in Google Scholar PubMed PubMed Central
Maticzka, D., Lange, S.J., Costa, F., and Backofen, R. (2014). GraphProt: modeling binding preferences of RNA-binding proteins. Genome Biol. 15: R17, https://doi.org/10.1186/gb-2014-15-1-r17.Search in Google Scholar PubMed PubMed Central
McCarthy, D.J., Chen, Y., and Smyth, G.K. (2012). Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 40: 4288–4297, https://doi.org/10.1093/nar/gks042.Search in Google Scholar PubMed PubMed Central
Navarro, S., Marinelli, P., Diaz-Caballero, M., and Ventura, S. (2015). The prion-like RNA-processing protein HNRPDL forms inherently toxic amyloid-like inclusion bodies in bacteria. Microb. Cell Factories 14: 102, https://doi.org/10.1186/s12934-015-0284-7.Search in Google Scholar PubMed PubMed Central
Plass, M., Rasmussen, S.H., and Krogh, A. (2017). Highly accessible AU-rich regions in 3′ untranslated regions are hotspots for binding of regulatory factors. PLOS Comput. Biol. 13: e1005460, https://doi.org/10.1371/journal.pcbi.1005460.Search in Google Scholar PubMed PubMed Central
Rahman, M.A., Masuda, A., Ohe, K., Ito, M., Hutchinson, D.O., Mayeda, A., Engel, A.G., and Ohno, K. (2013). HnRNP L and hnRNP LL antagonistically modulate PTB-mediated splicing suppression of CHRNA1 pre-mRNA. Sci. Rep. 3: 2931, https://doi.org/10.1038/srep02931.Search in Google Scholar PubMed PubMed Central
Ray, D., Kazan, H., Cook, K.B., Weirauch, M.T., Najafabadi, H.S., Li, X., Gueroussov, S., Albu, M., Zheng, H., Yang, A., et al.. (2013). A compendium of RNA-binding motifs for decoding gene regulation. Nature 499: 172–177, https://doi.org/10.1038/nature12311.Search in Google Scholar PubMed PubMed Central
Schaks, M., Giannone, G., and Rottner, K. (2019). Actin dynamics in cell migration. Essays Biochem. 63: 483–495, https://doi.org/10.1042/ebc20190015.Search in Google Scholar PubMed PubMed Central
Sheng, Z.-G. and Chen, M.-H. (2020). TPM4 aggravates the malignant progression of hepatocellular carcinoma through negatively regulating SUSD2. Eur. Rev. Med. Pharmacol. Sci. 24: 4756–4765, https://doi.org/10.26355/eurrev_202005_21164.Search in Google Scholar PubMed
Smith, S.A., Ray, D., Cook, K.B., Mallory, M.J., Hughes, T.R., and Lynch, K.W. (2013). Paralogs hnRNP L and hnRNP LL exhibit overlapping but distinct RNA binding constraints. PLoS ONE 8: e80701, https://doi.org/10.1371/journal.pone.0080701.Search in Google Scholar PubMed PubMed Central
Spellman, R., Llorian, M., and Smith, C.W.J. (2007). Crossregulation and functional redundancy between the splicing regulator PTB and its paralogs nPTB and ROD1. Mol. Cell 27: 420–434, https://doi.org/10.1016/j.molcel.2007.06.016.Search in Google Scholar PubMed PubMed Central
Sudhakaran, M. and Doseff, A.I. (2023). Role of heterogeneous nuclear ribonucleoproteins in the cancer-immune landscape. Int. J. Mol. Sci. 24: 5086, https://doi.org/10.3390/ijms24065086.Search in Google Scholar PubMed PubMed Central
Sun, Y., Chen, H., Lu, Y., Duo, J., Lei, L., OuYang, Y., Hao, Y., Da, Y., and Shen, X.-M. (2019). Limb girdle muscular dystrophy D3 HNRNPDL related in a Chinese family with distal muscle weakness caused by a mutation in the prion-like domain. J. Neurol. 266: 498–506, https://doi.org/10.1007/s00415-018-9165-4.Search in Google Scholar PubMed
Tchakarska, G. and Sola, B. (2020). The double dealing of cyclin D1. Cell Cycle 19: 163–178, https://doi.org/10.1080/15384101.2019.1706903.Search in Google Scholar PubMed PubMed Central
Tian, X.-Y., Li, J., Liu, T.-H., Li, D.-N., Wang, J.-J., Zhang, H., Deng, Z.-L., Chen, F.-J., and Cai, J.-P. (2020). The overexpression of AUF1 in colorectal cancer predicts a poor prognosis and promotes cancer progression by activating ERK and AKT pathways. Cancer Med. 9: 8612–8623, https://doi.org/10.1002/cam4.3464.Search in Google Scholar PubMed PubMed Central
Van Nostrand, E.L., Freese, P., Pratt, G.A., Wang, X., Wei, X., Xiao, R., Blue, S.M., Chen, J.-Y., Cody, N.A.L., Dominguez, D., et al.. (2020). A large-scale binding and functional map of human RNA-binding proteins. Nature 583: 711–719, https://doi.org/10.1038/s41586-020-2077-3.Search in Google Scholar PubMed PubMed Central
Vieira, N.M., Naslavsky, M.S., Licinio, L., Kok, F., Schlesinger, D., Vainzof, M., Sanchez, N., Kitajima, J.P., Gal, L., Cavaçana, N., et al.. (2014). A defect in the RNA-processing protein HNRPDL causes limb-girdle muscular dystrophy 1G (LGMD1G). Hum. Mol. Genet. 23: 4103–4110, https://doi.org/10.1093/hmg/ddu127.Search in Google Scholar PubMed
Vrakas, C.N., Herman, A.B., Ray, M., Kelemen, S.E., Scalia, R., and Autieri, M.V. (2019). RNA stability protein ILF3 mediates cytokine-induced angiogenesis. FASEB J. 33: 3304–3316, https://doi.org/10.1096/fj.201801315r.Search in Google Scholar
Vuong, J.K., Lin, C.-H., Zhang, M., Chen, L., Black, D.L., and Zheng, S. (2016). PTBP1 and PTBP2 serve both specific and redundant functions in neuronal pre-mRNA splicing. Cell Rep. 17: 2766–2775, https://doi.org/10.1016/j.celrep.2016.11.034.Search in Google Scholar PubMed PubMed Central
Wang, C., Zheng, M., Wang, S., Nie, X., Guo, Q., Gao, L., Li, X., Qi, Y., Liu, J., and Lin, B. (2019). Whole genome analysis and prognostic model construction based on alternative splicing events in endometrial cancer. BioMed Res. Int. 2019: 2686875, https://doi.org/10.1155/2019/2686875.Search in Google Scholar PubMed PubMed Central
Weigand, J.E., Boeckel, J.-N., Gellert, P., and Dimmeler, S. (2012). Hypoxia-induced alternative splicing in endothelial cells. PLOS ONE 7: e42697, https://doi.org/10.1371/journal.pone.0042697.Search in Google Scholar PubMed PubMed Central
White, E.J.F., Matsangos, A.E., and Wilson, G.M. (2017). AUF1 regulation of coding and noncoding RNA. WIREs RNA 8: e1393, https://doi.org/10.1002/wrna.1393.Search in Google Scholar PubMed PubMed Central
Wilks, C., Zheng, S.C., Chen, F.Y., Charles, R., Solomon, B., Ling, J.P., Imada, E.L., Zhang, D., Joseph, L., Leek, J.T., et al.. (2021). recount3: summaries and queries for large-scale RNA-seq expression and splicing. Genome Biol. 22: 323, https://doi.org/10.1186/s13059-021-02533-6.Search in Google Scholar PubMed PubMed Central
Wu, Y.-Y., Li, H., Lv, X.-Y., Wei, Q., Li, X., Liu, X.-Y., Zhou, Q., and Wei, Y.-Q. (2008). Overexpression of JKTBP1 induces androgen-independent LNCaP cell proliferation through activation of epidermal growth factor-receptor (EGF-R). Cell Biochem. Funct. 26: 467–477, https://doi.org/10.1002/cbf.1468.Search in Google Scholar PubMed
Yancopoulos, G.D., Davis, S., Gale, N.W., Rudge, J.S., Wiegand, S.J., and Holash, J. (2000). Vascular-specific growth factors and blood vessel formation. Nature 407: 242–248, https://doi.org/10.1038/35025215.Search in Google Scholar PubMed
Yang, R., Zheng, G., Ren, D., Chen, C., Zeng, C., Lu, W., and Li, H. (2018). The clinical significance and biological function of tropomyosin 4 in colon cancer. Biomed. Pharmacother. 101: 1–7, https://doi.org/10.1016/j.biopha.2018.01.166.Search in Google Scholar PubMed
Yoneda, R., Ueda, N., Uranishi, K., Hirasaki, M., and Kurokawa, R. (2020). Long noncoding RNA pncRNA-D reduces cyclin D1 gene expression and arrests cell cycle through RNA m6A modification. J. Biol. Chem. 295: 5626–5639, https://doi.org/10.1074/jbc.ra119.011556.Search in Google Scholar PubMed PubMed Central
Yoon, J.-H., De, S., Srikantan, S., Abdelmohsen, K., Grammatikakis, I., Kim, J., Kim, K.M., Noh, J.H., White, E.J.F., Martindale, J.L., et al.. (2014). PAR-CLIP analysis uncovers AUF1 impact on target RNA fate and genome integrity. Nat. Commun. 5: 5248, https://doi.org/10.1038/ncomms6248.Search in Google Scholar PubMed PubMed Central
Yu, B. and Wang, S. (2018). Angio-LncRs: LncRNAs that regulate angiogenesis and vascular disease. Theranostics 8: 3654–3675, https://doi.org/10.7150/thno.26024.Search in Google Scholar PubMed PubMed Central
Yu, G., Wang, L.-G., Han, Y., and He, Q.-Y. (2012). clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS J. Integr. Biol. 16: 284–287, https://doi.org/10.1089/omi.2011.0118.Search in Google Scholar PubMed PubMed Central
Zhang, P., Ji, D., Hu, X., Ni, H., Ma, W., Zhang, X., Liao, S., Zeng, Z., Zhao, Y., and Zhou, H. (2018). Oncogenic heterogeneous nuclear ribonucleoprotein D-like promotes the growth of human colon cancer SW620 cells via its regulation of cell-cycle. Acta Biochim. Biophys. Sin. 50: 880–887, https://doi.org/10.1093/abbs/gmy085.Search in Google Scholar PubMed
Zhao, X., Jiang, M., and Wang, Z. (2019). TPM4 promotes cell migration by modulating F-actin formation in lung cancer. OncoTargets Ther. 12: 4055–4063, https://doi.org/10.2147/ott.s198542.Search in Google Scholar
Zhou, H., Ge, Y., Sun, L., Ma, W., Wu, J., Zhang, X., Hu, X., Eaves, C.J., Wu, D., and Zhao, Y. (2014). Growth arrest specific 2 is up-regulated in chronic myeloid leukemia cells and required for their growth. PLOS ONE 9: e86195, https://doi.org/10.1371/journal.pone.0086195.Search in Google Scholar PubMed PubMed Central
Zhu, A., Ibrahim, J.G., and Love, M.I. (2019). Heavy-tailed prior distributions for sequence count data: removing the noise and preserving large differences. Bioinformatics 35: 2084–2092, https://doi.org/10.1093/bioinformatics/bty895.Search in Google Scholar PubMed PubMed Central
Supplementary Material
This article contains supplementary material (https://doi.org/10.1515/hsz-2023-0254).
© 2023 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Research Articles/Short Communications
- Genes and Nucleic Acids
- Heterogenous nuclear ribonucleoprotein D-like controls endothelial cell functions
- Cell Biology and Signaling
- TIF1γ and SMAD4 regulation in colorectal cancer: impact on cell proliferation and liver metastasis
- Methionine restriction attenuates the migration and invasion of gastric cancer cells by inhibiting nuclear p65 translocation through TRIM47
- Celastrol inhibits angiogenesis and the biological processes of MDA-MB-231 cells via the DEGS1/S1P signaling pathway
- Proteolysis
- Cathepsin L-mediated EGFR cleavage affects intracellular signalling pathways in cancer
Articles in the same Issue
- Frontmatter
- Research Articles/Short Communications
- Genes and Nucleic Acids
- Heterogenous nuclear ribonucleoprotein D-like controls endothelial cell functions
- Cell Biology and Signaling
- TIF1γ and SMAD4 regulation in colorectal cancer: impact on cell proliferation and liver metastasis
- Methionine restriction attenuates the migration and invasion of gastric cancer cells by inhibiting nuclear p65 translocation through TRIM47
- Celastrol inhibits angiogenesis and the biological processes of MDA-MB-231 cells via the DEGS1/S1P signaling pathway
- Proteolysis
- Cathepsin L-mediated EGFR cleavage affects intracellular signalling pathways in cancer