Figure 4 - uploaded by Chunli Yan
Content may be subject to copyright.
Constitutive enhancer activity of AcH3 peak sequences at 8q24. The DNA sequence containing each of the 15 identified AcH3 sites or a control sequence from the neighboring unacetylated region was inserted upstream of TK-luciferase reporter vector. The constructs were transfected into 5 different cell lines (LNCaP, PC3, HCT 115, COLO 205, and MCF7) along with pRL-TK Renilla luciferase plasmid for 24 h. Dual luciferase assays were conducted. The results were normalized against the internal Renilla control for each transfection. The luciferase activity of the control region was defined as 1. Relative luciferase activity values are presented as mean 6 SD of triplicate transfections. doi:10.1371/journal.pgen.1000597.g004 

Constitutive enhancer activity of AcH3 peak sequences at 8q24. The DNA sequence containing each of the 15 identified AcH3 sites or a control sequence from the neighboring unacetylated region was inserted upstream of TK-luciferase reporter vector. The constructs were transfected into 5 different cell lines (LNCaP, PC3, HCT 115, COLO 205, and MCF7) along with pRL-TK Renilla luciferase plasmid for 24 h. Dual luciferase assays were conducted. The results were normalized against the internal Renilla control for each transfection. The luciferase activity of the control region was defined as 1. Relative luciferase activity values are presented as mean 6 SD of triplicate transfections. doi:10.1371/journal.pgen.1000597.g004 

Source publication
Article
Full-text available
Multiple discrete regions at 8q24 were recently shown to contain alleles that predispose to many cancers including prostate, breast, and colon. These regions are far from any annotated gene and their biological activities have been unknown. Here we profiled a 5-megabase chromatin segment encompassing all the risk regions for RNA expression, histone...

Context in source publication

Context 1
... contrast, region 1 was totally devoid of transcripts in all tissues and cell lines. We did not investigate the transcripts originating in region 2 and 3 further, since their abundance in prostate tissue was not affected by risk haplotypes in the region [14]. In parallel, we generated high-resolution epigenomic profiles for the entire 5-Mb interval using ChIP-chip. For this purpose we hybridized ChIP material to the same tiling array used earlier for transcriptional profiling. Initially, we analyzed AcH3 in three cell lines representing prostate (LNCaP), breast (MCF7) and colorectal (HCT116) cancer. Because three regions independently impose prostate cancer risk, we also interrogated two prostate cancer cell lines (PC3 & LNCaP) more extensively for other key epigenetic marks at high resolution. Additional histone modifications chosen were the activation marks H3K4me1 & H3K4me3 [10], the transcription elongation mark H3K36me3 and the polycomb repressive mark H3K27me3. We also profiled RNA polymerase II (RNAPII) and patterns of androgen receptor (AR) occupied regions (ARORs). This entire multi-dimensional dataset (including cDNA profiles) was then subjected to extensive statistical analysis using spatial clustering, a new method that allows the dissection of large genomic regions into distinct clusters, each reflecting a specific combinatorial pattern of epigenetic marks in an unbiased manner [15,16]. Spatial clustering of the 5-Mb region surrounding and including the 8q24 risk loci is shown in Figure 2A and 2B. This unsupervised cluster analysis revealed domains of combinatorial histone modification and cDNA patterns, and determined the most likely type of behavior at each genomic locus. Six domain types were evident, color-coded and numbered I–VI (Figure 2A). The cancer risk regions are bordered by two distinct domains located 2-Mb apart: a 1-Mb type IV domain (located , 127 Mb), which is weakly enriched with H3K27me3 marks, and a type I domain- encompassing MYC (located , 129 Mb), which is strongly enriched with activation-associated marks and transcription (Figure 2B). The prostate cancer risk regions 1–3 were assigned to a type VI domain, indicating that the chromatin of the risk- linked domain is uniquely structured, and includes features that are distinctly different from the aforementioned flanking regions. Importantly, an additional LNCaP H3K27me3 domain (domain IV) is located downstream of MYC, with significant H3K27me3 enrichment limited to LNCaP (Figure S3). As H3K27me3 is a modification associated with polycomb-mediated repression, this suggests that in LNCaP the chromosomal architecture may group the MYC genes and the risk regions in between large repressed domains, possibly facilitating interactions between them. A higher-resolution epigenetic map of the risk regions in LNCaP is shown in Figure 3. As noted above, regions 1 and 3 were not robustly transcribed in either the normal tissues or prostate cancer cell lines. The histones in this region, however, were highly modified in LNCaP, with particular enrichment for active chromatin marks, i.e. AcH3, H3K4me1 and H3K4me3. Addi- tionally we observed significant occupancy of AR and RNAPII. Importantly, these patterns of activity were absent from PC3, which does not express the AR. The risk regions were also enriched for the elongation mark H3K36me3; however, in line with the general lack of transcription, the H3K36me3 areas were not polarized to a specific side of adjacent RNAPII peaks. Risk region 1 included, in addition, the three strongest H3K27me3 peaks in the 5-Mb region, suggesting that some polycomb dependent repression may affect region 1 activity in LNCaP cells. The epigenomic organization of the risk regions therefore reflects multiple hotspots of active chromatin, involving RNAPII, AR occupancy and activation as well as elongation marks, but without any detectable transcriptional footprints. Thus, these features may be understood as describing enhancers that regulate either dormant transcriptional units in cis or remote active transcriptional units in trans . We note that we could not rule out the possibility of small non-coding RNAs being transcribed from the region, since RNA species shorter than 200-bp were excluded from our preparation. In order to investigate the regulatory potential of the loci exhibiting active chromatin marks, we next performed a systematic series of heterologous enhancer assays, focusing initially on defined acetylation peaks contained within the cancer risk intervals (called AcP1 through AcP15, in Figure 3). We cloned approximately 1.5-kb DNA fragments, centered on AcPs from LNCaP, HCT116 or MCF7 cells, upstream of a luciferase reporter gene driven by the thymidine kinase ( TK ) minimal promoter. Enhancer activities of the fragments were determined by transient transfection and luciferase assays in LNCaP & PC3 (prostate cancer cells), HCT116 & COLO 205 (colorectal cancer cells) and MCF7 (breast cancer cells) (Figure 4). AcP6 (in the breast cancer risk region) and AcP10 (in prostate cancer/colorectal cancer risk region 3) had the most pronounced enhancer activities, whereas AcPs12 – 15 (in prostate cancer risk region 1) had activities that were lower, but clear compared to the negative control and several other AcPs. Interestingly, these active enhancers also displayed unmistakable H3K4me1 and H3K4me3 marks. The results suggest that some of the active chromatin foci we identified (Figure 3, right inset) have intrinsic enhancer activities within cellular contexts. This concept was further supported in a parallel study, in colorectal cells, which demonstrated that region 3, encompassing AcP10 and harboring SNP rs6983267, bound transcription factor T-cell factor 4 (TCF4) in an allele specific manner [17]. In the present study we did not study this region but rather analyzed region 1 further in prostate cancer cells. Risk region 1 is specifically linked to prostate cancer risk, and the three most robust acetylation peaks also exhibited strong AR binding (Figure 3). This region additionally exhibited both active marks (H3K4me1&3, found at active TSSs and enhancers) and inactive marks (H3K27me3, found throughout silenced genes and some intergenic regions), as well as occupancy of RNAPII. Further analysis of potential AR-mediated enhancers was strongly justified considering the major involvement of AR in all phases of prostate cancer development, including advanced ablation-resistant disease [18]. Consequently, we investigated the potential for androgen- dependent enhancer activities in this region. First, to verify and further characterize the AR binding at AcPs13, -14 & -15 as suggested by ChIP-chip (Figure 3), site-specific ChIP analyses were conducted using cells treated with dihydrotestosterone (DHT) or vehicle (Figure 5A). All three sites, in particular AcPs 14 & 15, revealed strong DHT-stimulated AR occupancy. Second, to test directly for androgen-dependent enhancer activities, we cloned narrower regions ( , 0.5-kb fragments) than the original AcP regions (which were , 1.5-kb in length), centered around the AR occupancy peaks in the same TK-luciferase reporter plasmid described above, named AROR13, -14 & -15, respectively. LNCaP cells, which express AR, were transfected with these plasmids and luciferase activity was measured. The results revealed robust DHT-dependent enhancer activity in AROR14 and -15, even higher than that of the PSA enhancer used as a positive control (Figure 5B), and this level of activity roughly correlated with the DHT-stimulated AR occupancies at the respective sites (Figure 5A). AROR-14 exhibited a remarkable basal activity but only a 3-fold response to DHT. In order to capture all common genetic variations in this region, we resequenced ARORs14 and -15 in prostate cancer cases of European ancestry (172 chromosomes). Through this effort we identified two SNPs in AROR15 that were strongly correlated with the risk variant rs10090154 (reported in [4]), which itself was not located within an AROR (rs11986220, r 2 = 1.0 and rs11988857, r 2 = 0.923; Figure 5C). We introduced all allelic combinations of both SNPs into the AROR15 reporter, creating 4 plasmids representative of the 4 alleles as shown in Figure 5D. In six independent experiments, using six independently constructed sets of plasmids, the DHT-dependent enhancer activity observed with the A-allele of rs11986220 was , 2-fold higher than the enhancer activity observed with the T allele, regardless of the SNP at rs11988857. Since the A allele at rs11986220 is also the allele associated with the risk allele for prostate cancer at rs10090154, these results suggest that the increased androgen-mediated activity of the enhancer may upregulate expression of an important oncogene in prostate epithelial cells. What are the mechanism(s) that govern the SNP effect on the DHT-mediated enhancer activity described above? Interestingly, the SNP at rs11986220 resides within a putative binding site for forkhead transcription factors, with the A allele better matching the consensus sequence (Figure S4). An interesting and relevant forkhead transcription factor is FoxA1, which has been implicated in augmenting responsiveness of some ARORs to androgens [12,19]. Although LNCaP cells are homozygous for the T allele at rs11986220, the physical presence of FoxA1 at the AROR15 enhancer was nevertheless demonstrated by site-specific ChIP analysis (Figure 5E). Importantly, this occupancy was enhanced by DHT treatment of the cells. In a competition electromobility shift assay (Figure 5F), an oligonucleotide centered around SNP rs11986220 competed better for FoxA1 binding to a consensus Fox oligonucleotide, when the SNP position was an A as compared to a T. Thus, the stronger DHT-responsiveness of the AROR15 enhancer observed with the A SNP at rs11986220 is attributable to higher affinity for the AR collaborator, FoxA1. Since the histone acetyl transferase and ...

Citations

... Notably, mutations in FOXA1 enhancers were previously shown to alter TF bindings in primary prostate tumors 34 . And, also in line with our observations, FOXA1 enhancer region has been previously reported to be coupled to MYC 40 and has been shown to have a strong binding of AR 41 . ...
Article
Full-text available
We introduce Promoter-Enhancer-Guided Interaction Networks (PENGUIN), a method for studying protein-protein interaction (PPI) networks within enhancer-promoter interactions. PENGUIN integrates H3K27ac-HiChIP data with tissue-specific PPIs to define enhancer-promoter PPI networks (EPINs). We validated PENGUIN using cancer (LNCaP) and benign (LHSAR) prostate cell lines. Our analysis detected EPIN clusters enriched with the architectural protein CTCF, a regulator of enhancer-promoter interactions. CTCF presence was coupled with the prevalence of prostate cancer (PrCa) single nucleotide polymorphisms (SNPs) within the same EPIN clusters, suggesting functional implications in PrCa. Within the EPINs displaying enrichments in both CTCF and PrCa SNPs, we also show enrichment in oncogenes. We substantiated our identified SNPs through CRISPR/Cas9 knockout and RNAi screens experiments. Here we show that PENGUIN provides insights into the intricate interplay between enhancer-promoter interactions and PPI networks, which are crucial for identifying key genes and potential intervention targets. A dedicated server is available at https://penguin.life.bsc.es/.
... Significantly, AR and MYC pathways are inversely paired in PCa cells due to co-factor competition; and their partial signature compensation would safeguard essential transcriptional programs and cellular activities during functional transition. The sensitivity of the MYC gene to transcriptional regulation is intensified by its short halflife in both mRNA and proteins and its distal regulation by SEs that are vulnerable to environmental perturbation [3,[18][19][20][21]33]. Taken advantages of their androgen hyper-sensitivity, MYC and the 8q-24 locus were singled out in current report to validate AR bi-directional regulation. ...
Article
Full-text available
Background Androgen receptor (AR) activation and repression dual-functionality only became known recently and still remains intriguing in prostate cancer (PCa). MYC is a prominent oncogene that functionally entangles with AR signaling in PCa. Further exploration of AR regulatory mechanisms on MYC gene transcription bears clinical and translation significance. Methods Bioinformatics analysis of PCa cell line and clinical RNA-Seq and ChIP-Seq (chromatin immunoprecipitation-sequencing) datasets to anchor interactions of AR and MYC transcriptional networks. ChIP-qPCR and 3C (chromosome conformation capture) analyses to probe MYC distal regulation by AR binding sites (ABSs). CRISPR/Cas9-mediated genome-editing to specify functions of ABS within the 8q24-MYC locus on androgen-mediated MYC transcription. Global FoxA1 and HoxB13 distribution profiling to advance AR transcriptional mechanisms. Results Here we recognize AR bi-directional transcription mechanisms by exploiting the prominent 8q24-MYC locus conferring androgen hyper-sensitivity. At ~ 25 Kb downstream of the MYC gene, we identified an undefined ABS, P10. By chromatin analyses, we validated androgen-dependent spatial interaction between P10 and MYC-Promoter (MYC-Pro) and temporal epigenetic repression of these MYC-proximal elements. We next designed a CRISPR/Cas9-mediated double genomic knock-out (KO) strategy to show that P10-KO slightly lessened androgen-elicited MYC transrepression in LNCaP-AR cells. In similar genomic editing assays, androgen-mediated MYC repression became slightly deepened upon KO of P11, an ABS in the PVT1 gene locus highly enriched in AR-binding motifs and peaks. We also investigated multiple ABSs in the established PCAT1 super-enhancer that distally interacts with MYC-Pro for transactivation, with each KO pool consistently shown to relieve androgen-elicited MYC repression. In the end, we systemically assessed androgen effects in the 8q24-MYC locus and along PCa genome to generalize H3K27ac and BRD4 re-distribution from pioneer factors (FoxA1 and HoxB13) to AR sites. Conclusion Together, we reconciled these observations by unifying AR dual-functions that are mechanistically coupled to and equilibrated by co-factor redistribution. Graphical Abstract
... Certain PCa risk variants such as rs6983267 or rs7008482 are constitutive alleles in the African ancestry population and largely explain the 1.5-fold increased risk of PCa in this population. Functional regions in 8q24 related to PCa risk have been mainly linked to MYC regulation and the androgen pathway through FOXA1 or transcription factor 7-like 2 (TCF7L2) binding to AR binding sites [22]. ...
Article
Full-text available
Objective To review the current status of germline and somatic (tumour) genetic testing for prostate cancer (PCa), and its relevance for clinical practice. Methods A narrative synthesis of various molecular profiles related to their clinical context was carried out. Current guidelines for genetic testing and its feasibility in clinical practice were analysed. We report the main identified genetic sequencing results or functional genomic scores for PCa published in the literature or obtained from the French PROGENE study. Results The molecular alterations observed in PCa are mostly linked to disruption of the androgen receptor (AR) pathway or DNA repair deficiency. The main known germline mutations affect the BReast CAncer gene 2 (BRCA2) and homeobox B13 (HOXB13) genes, whereas AR and tumour protein p53 (TP53) are the genes with most frequent somatic alterations in tumours from men with metastatic PCa. Molecular tests are now available for detecting some of these germline or somatic alterations and sometimes recommended by guidelines, but their utilisation must combine rationality and feasibility. They can guide specific therapies, notably for the management of metastatic disease. Indeed, following androgen deprivation, targeted therapies for PCa currently include poly‐(ADP‐ribose)‐polymerase (PARP) inhibitors, immune checkpoint inhibitors, and prostate‐specific membrane antigen (PSMA)‐guided radiotherapy. The genetic tests currently approved for targeted therapies remain limited to the detection of BRCA1 and BRCA2 mutation and DNA mismatch repair deficiency, while large panels are recommended for germline analyses, not only for inherited cancer predisposing syndrome, but also for metastatic PCa. Conclusions Further consensus aligning germline with somatic molecular analysis in metastatic PCa is required, including genomics scars, emergent immunohistochemistry, or functional pre‐screen imaging. With rapid advances in knowledge and technology in the field, continuous updating of guidelines to help the clinical management of these individuals, and well‐conducted studies to evaluate the benefits of genetic testing are needed.
... Signi cantly, AR and MYC pathways are inversely paired in PCa cells due to co-factor competition; and their partial functional compensation would safeguard essential transcriptional programs and cellular activities. The sensitivity of the MYC gene to transcriptional regulation is intensi ed by its short half-lives in both mRNA and proteins and its distal regulation by SEs that are vulnerable to environmental perturbation [3,[18][19][20][21]25]. Taken advantages of their androgen hyper-sensitivity, MYC and the 8q-24 locus were singled out in current report to validate AR bi-directional regulation. ...
Preprint
Full-text available
Background:Androgen receptor (AR) activation and repression dual-functionality only becomes known recently and still remains intriguing in prostate cancer (PCa). MYC is a prominent oncogene functionally entangled with AR signaling in PCa. However, AR regulatory mechanisms on MYC gene transcription remains unclear. Methods:Bioinformatics analysis of androgen-mediated RNA-Seq and MYC ChIP-Seq datasets are used for AR and MYC transcriptional networks. AR ChIP-qPCR analysis are programed to find AR binding sites (ABSs) which regulate MYC transcription. 3C-qPCR and 3C-ddPCR analyses affirmed androgen-dependent MYC-Pro-P10 interaction. CRISPR/Cas9-mediated double genomic knock-out (KO) strategy is used to show that P10-KO slightly lessened androgen-elicited MYC transrepression. Results:Here we recognize AR bi-directional transcription mechanisms by exploiting the prominent 8q24-MYC locus conferring androgen hyper-sensitivity. At ~25Kb downstream of the MYC gene, we identified an undefined ABS, P10. By chromatin analyses, we validated androgen-dependent spatial interaction between P10 and MYC-Promoter (MYC-Pro) and temporal epigenetic repression of these MYC-proximal elements. We next designed a CRISPR/Cas9-mediated double genomic knock-out (KO) strategy to show that P10-KO slightly lessened androgen-elicited MYC transrepression in LNCaP-AR cells. In similar genomic editing assays, androgen-mediated MYC repression became slightly deepened upon KO of P11, an ABS in the PVT1 gene locus highly enriched in AR-binding motifs and peaks. We also investigated multiple ABSs in the established PCAT1 super-enhancer that distally interacts with MYC-Pro for transactivation, with each KO pool consistently shown to relieve androgen-elicited MYC repression. In the end, we systemically assessed androgen effects in the 8q24-MYC locus and along PCa genome to generalize H3K27ac and BRD4 re-distribution from pioneer factors (FoxA1 and HoxB13) to AR sites. Conclusion:Together, we reconciled these observations by unifying AR dual-functions that are mechanistically coupled to and equilibrated by co-factor redistribution.
... The amplification of the chromosome 8q24 region, where the CanCord34 genes reside, is one of the most common genomic events in multiple cancer types [7][8][9][10][11]. We next determined whether the CanCord34 genes are co-overexpressed in other cancer types in addition to breast cancer. ...
Article
Full-text available
Many human cancers, including breast cancer, are polygenic and involve the co-dysregulation of multiple regulatory molecules and pathways. Though the overexpression of genes and amplified chromosomal regions have been closely linked in breast cancer, the notion of the co-upregulation of genes at a single locus remains poorly described. Here, we describe the co-overexpression of 34 continuously organized protein-coding genes with diverse functions at 8q.24.3(143437655-144326919) in breast and other cancer types, the CanCord34 genes. In total, 10 out of 34 genes have not been reported to be overexpressed in breast cancer. Interestingly, the overexpression of CanCord34 genes is not necessarily associated with genomic amplification and is independent of hormonal or HER2 status in breast cancer. CanCord34 genes exhibit diverse known and predicted functions, including enzymatic activities, cell viability, multipotency, cancer stem cells, and secretory activities, including extracellular vesicles. The co-overexpression of 33 of the CanCord34 genes in a multivariant analysis was correlated with poor survival among patients with breast cancer. The analysis of the genome-wide RNAi functional screening, cell dependency fitness, and breast cancer stem cell databases indicated that three diverse overexpressed CanCord34 genes, including a component of spliceosome PUF60, a component of exosome complex EXOSC4, and a ribosomal biogenesis factor BOP1, shared roles in cell viability, cell fitness, and stem cell phenotypes. In addition, 17 of the CanCord34 genes were found in the microvesicles (MVs) secreted from the mesenchymal stem cells that were primed with MDA-MB-231 breast cancer cells. Since these MVs were important in the chemoresistance and dedifferentiation of breast cancer cells into cancer stem cells, these findings highlight the significance of the CanCord34 genes in cellular communications. In brief, the persistent co-overexpression of CanCord34 genes with diverse functions can lead to the dysregulation of complementary functions in breast cancer. In brief, the present study provides new insights into the polygenic nature of breast cancer and opens new research avenues for basic, preclinical, and therapeutic studies in human cancer.
... Zhou et al. 2020). And, also in line with our observations, FOXA1 enhancer region has been previously reported to be coupled to MYC (Sur et al. 2013) and has been shown to have a strong binding of AR (Jia et al. 2009). ...
Preprint
Full-text available
Here we introduce Promoter-ENhancer-GUided Interaction Networks (PENGUIN), a method to uncover protein-protein interaction (PPI) networks at enhancer-promoter contacts. By integrating H3K27ac-HiChIP data and tissue-specific PPI information, PENGUIN enables cluster enhancers-promoter PPI networks (EPINs) and pinpoint actionable factors. Validating PENGUIN in cancer (LNCaP) and benign (LHSAR) prostate cell lines, we observed distinct CTCF-enriched clusters, which identifies diverse chromatin conformations. In LNCaP, we found an EPIN cluster enriched with oncogenes and prostate cancer-associated SNPs. We uncovered a total of 208 SNPs in LNCaP EPINs and used CRISPR/Cas9 knockout and RNAi screens to confirm their relevance. PENGUIN’s application in prostate cancer demonstrates its potential for studying human diseases. The approach allows exploration in different cell types and combinations of GWAS data, offering promising avenues for future investigations. In conclusion, PENGUIN provides valuable insights into the interplay between enhancer-promoter interactions and PPI networks, facilitating the identification of relevant genes and potential intervention targets.
... According to recent studies, eRNAs are highly engaged in oncogene activation and aberrant signaling pathways, which contribute to tumorigenesis [45]. The eRNAs transcribed from the super-enhancer regions on 8q24 serve as a good example of how this works [30,[63][64][65][66]. Using the chromosome conformation capture (3C) technique, researchers have found long-range physical interactions between eRNAs from 8q24 and MYC, whose dysregulation is a significant symbol of multiple cancer types [63,[67][68][69][70][71]. For example, colon cancer-associated transcript 1 (CCAT1), one well-studied eRNA transcribed from the 8q24 locus, can be activated through long-range interaction with MYC in many cancers, such as colorectal cancer (CRC), prostate adenocarcinoma (PRAD), breast invasive carcinoma (BRCA) and esophageal carcinoma (ESCA) [63,[72][73][74]. ...
... These pathways are vital to tumorigenesis by contributing to proliferation, differentiation and anti-apoptosis [80,81]. Taken as a whole, the variants, modifications or abnormal activation of 8q24 regions can induce overexpression of its transcribed eRNAs, which then trigger the pathogenesis of multiple human cancers, including CRC, PRAD, BRCA, ESCA and hematological malignancies [65,66,[69][70][71]82]. The interaction of eRNAs with Bromodomain and Extraterminal (BET) proteins also showed some insights into cancer development. ...
Article
Full-text available
Enhancer RNAs (eRNAs), a class of non-coding RNAs (ncRNAs) transcribed from enhancer regions, serve as a type of critical regulatory element in gene expression. There is increasing evidence demonstrating that the aberrant expression of eRNAs can be broadly detected in various human diseases. Some studies also revealed the potential clinical utility of eRNAs in these diseases. In this review, we summarized the recent studies regarding the pathological mechanisms of eRNAs as well as their potential utility across human diseases, including cancers, neurodegenerative disorders, cardiovascular diseases and metabolic diseases. It could help us to understand how eRNAs are engaged in the processes of diseases and to obtain better insight of eRNAs in diagnosis, prognosis or therapy. The studies we reviewed here indicate the enormous therapeutic potency of eRNAs across human diseases.
... The 8q24 locus is a highly conserved genomic region traditionally referred as a "gene desert", devoid of transcriptional activity. However, it contains functional regulatory elements, particularly enhancers [10,11]. Fine mapping of this locus revealed five clusters of multiple independent variants, showing strong linkage disequilibrium (LD), and association with susceptibility to different types of cancer [12][13][14][15]. ...
Article
Full-text available
Variants of 8q24 locus have been associated with prostate cancer (PCa) susceptibility. This study aims to analyze the genetic basis of PCa susceptibility in Mexican men by analyzing SNPs in the 8q24 locus for the first time. A case-control study was performed in 875 men recruited from the Mexican Social Security Institute, 326 patients with PCa, and 549 non-PCa patients (88 with benign prostatic hyperplasia BPH and 461 healthy controls). The 8q24 locus SNPs: rs16901979, rs16983267, rs1447295, and rs7837328 were genotyped by allelic discrimination assays using TaqMan probes. Statistical analysis was performed using Epi Info statistical 7.0 and SNPstats softwares. All genotype frequencies were in Hardy-Weinberg Equilibrium. No differences were observed in genotype distribution between PCa and non-PCa patients for rs6983267. Under different inheritance models, the rs16901979, rs1447295, and rs7837328 SNPs were associated with PCa (OR = 2.8, 1.8, and 1.72, respectively; Pc < 0.001) when comparing PCa patients against controls. This association remains between PCa and BPH patients under different models (OR = 8.5, 2.2, and 1.9, respectively; Pc < 0.001). There were no significant differences in allele and genotype distribution among BPH patients and controls. The combined effect of the alleles CGAA for the SNPs rs16901979, rs6983267, rs1447295, and rs7837328 showed significant differences between PCa patients and controls (OR = 2.9, 95% CI = 1.48–5.83, Pc = 0.008). Four 8q24 variants were not associated with D’Amico score, age at diagnosis, and bone metastases. Our study provides the first confirmation that variants rs16901979, rs1447295, and 7837328 at 8q24 locus are associated with PCa susceptibility in Mexican men.
... Fox proteins regulate the expression of genes involved in branching morphogenesis, brain development, axon guidance and maintaining stem-cell pluripotency, among a host of other biological processes (35,36). The FOXA1 motif is known to be particularly vulnerable to variants that disrupt the three consecutive adenines, which is true of rs356182 (Fig. 3A), and was previously shown to result in pathological consequences (37,38). The FOXO family has important roles in cell cycle, apoptosis, oxidative stress and differentiation (39). ...
Article
Full-text available
One of the most significant risk variants for Parkinson's disease (PD), rs356182, is located at the PD-associated locus near the alpha-synuclein (α-syn) encoding gene, SNCA. SNCA-proximal variants, including rs356182, are thought to function in PD risk through enhancers via allele-specific regulatory effects on SNCA expression. However, this interpretation discounts the complex activity of genetic enhancers and possible nonconical functions of α-syn. Here we investigated a novel risk mechanism for rs356182. We use CRISPR-Cas9 in LUHMES cells, a model for dopaminergic midbrain neurons, to generate precise hemizygous lesions at rs356182. The PD-protective (A/-), PD-risk (G/-), and wildtype (A/G) clones were neuronally differentiated and then compared transcriptionally and morphologically. Among the affected genes was SNCA, whose expression was promoted by the PD-protective allele (A) and repressed in its absence. In addition to SNCA, hundreds of genes were differentially expressed and associated with neurogenesis and axonogenesis- an effect not typically ascribed to α-syn. We also found that the transcription factor FOXO3 specifically binds to the rs356182 A-allele in differentiated LUHMES cells. Finally, we compared the results from the rs356182-edited cells to our previously published knockouts of SNCA and found only minimal overlap between the sets of significant differentially expressed genes. Together, the data implicate a risk mechanism for rs356182 in which the risk-allele (G) is associated with abnormal neuron development, independent of SNCA expression. We speculate that these pathological effects manifest as a diminished population of dopaminergic neurons during development leading to the predisposition for PD later in life.
... Given the prevalence of low-affinity binding sites within developmental enhancers, we hypothesize that single nucleotide variants (SNVs) that increase the affinity of binding sites could cause gain-offunction gene expression that contributes to phenotypic change. Variants that alter binding affinity have been linked to changes in gene expression and phenotypes in a handful of cases, however these are one off examples identified in an ad hoc way (Bond et al., 2004;French et al., 2013;Grant et al., 1996;Huang et al., 2014;Jia et al., 2009;Tuupanen et al., 2009). In contrast, here we investigate if searching for affinity-optimizing variants is a generalizable strategy to pinpoint enhancer variants that contribute to gain-of-function gene expression and phenotypic changes within the context of heart development. ...
... Changes in binding affinity have been linked to changes in gene expression and phenotypes in a handful of studies (Bond et al., 2004;French et al., 2013;Grant et al., 1996;Huang et al., 2014;Jia et al., 2009;Tuupanen et al., 2009). Notably, these are one off examples rather than systematic approaches to find causal variants across disparate enhancers, associated with different diseases and traits across species and cell types. ...
Preprint
Full-text available
Enhancers direct precise gene expression patterns during development and harbor the majority of variants associated with disease. We find that suboptimal affinity ETS transcription factor binding sites are prevalent within Ciona and human developmental heart enhancers. Here we demonstrate in two diverse systems, Ciona intestinalis and human iPSC-derived cardiomyocytes (iPSC-CMs), that single nucleotide changes can optimize the affinity of ETS binding sites, leading to gain-of-function gene expression associated with heart phenotypes. In Ciona , ETS affinity-optimizing SNVs lead to ectopic expression and phenotypic changes including two beating hearts. In human iPSC-CMs, an affinity-optimizing SNV associated with QRS duration occurs within an SCN5A enhancer and leads to increased enhancer activity. Our mechanistic approach provides a much-needed systematic framework that works across different enhancers, cell types and species to pinpoint causal enhancer variants contributing to enhanceropathies, phenotypic diversity and evolutionary changes. In Brief The prevalent use of low-affinity ETS sites within developmental heart enhancers creates vulnerability within genomes whereby single nucleotide changes can dramatically increase binding affinity, causing gain-of-function enhancer activity that impacts heart development. Highlights ETS affinity-optimizing SNVs can lead to migration defects and a multi-chambered heart. An ETS affinity-optimizing human SNV within an SCN5A enhancer increases expression and is associated with QRS duration. Searching for ETS affinity-optimizing variants is a systematic and generalizable approach to pinpoint causal enhancer variants.