Next Article in Journal
The Multifaceted Roles of CHROMR in Innate Immunity, Cancer, and Cholesterol Homeostasis
Previous Article in Journal
An Emphasis on the Role of Long Non-Coding RNAs in Viral Gene Expression, Pathogenesis, and Innate Immunity in Viral Chicken Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Human Papillomavirus-Encoded microRNAs as Regulators of Human Gene Expression in Anal Squamous Cell Carcinoma: A Meta-Transcriptomics Study

by
Daniel J. García
1,†,
Marco A. Pulpillo-Berrocal
1,†,
José L. Ruiz
1,2,
Eduardo Andrés-León
1,* and
Laura C. Terrón-Camero
1,*
1
Institute of Parasitology and Biomedicine “López-Neyra” (IPBLN), Spanish National Research Council (CSIC), 18016 Granada, Spain
2
Functional Genomics Center Zurich, ETH Zurich, University of Zurich, 8057 Zurich, Switzerland
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Non-Coding RNA 2025, 11(3), 43; https://doi.org/10.3390/ncrna11030043
Submission received: 11 March 2025 / Revised: 22 May 2025 / Accepted: 4 June 2025 / Published: 9 June 2025

Abstract

:
Introduction: Anal squamous cell carcinoma (ASCC) is a rare but increasingly common gastrointestinal malignancy, mainly associated with oncogenic human papillomaviruses (HPVs). The role of non-coding RNAs (ncRNAs) in tumorigenesis is recognized, but the impact of viral ncRNAs on host gene expression remains unclear. Methods: We re-analyzed total RNA-Seq data from 70 anal biopsies: 31 low-grade squamous intraepithelial lesions (LGSIL), 16 high-grade SIL (HGSIL), and 23 ASCC cases. Microbial composition was assessed taxonomically. Novel viral miRNAs were predicted using vsRNAfinder and linked to host targets using TargetScan and expression correlation analyses. Results: Microbial profiling revealed significant differences in abundance, with Alphapapillomaviruses types 9, 10, and 14 enriched across lesion grades. We identified 90 novel viral miRNAs and 177 significant anti-correlated miRNA–mRNA interactions. Target genes were enriched in pathways related to cell cycle, epithelial–mesenchymal transition, lipid metabolism, immune modulation, and viral replication. Discussion: Our findings suggest that HPV-derived miRNAs, including those from low-risk types, may contribute to neoplastic transformation by modulating host regulatory networks. Conclusion: This study highlights viral miRNAs as potential drivers of HPV-related anal cancer and supports their utility as early biomarkers and therapeutic targets in ASCC.

1. Introduction

The vast majority of the human genome (~98%) encodes RNAs that do not undergo translation into proteins at the ribosomes, collectively referred to as non-coding RNAs (ncRNAs) [1,2]. Since their identification as ‘junk RNA’, ncRNAs have gained attention for their crucial biological roles, driving a fundamental paradigm in RNA research. Although initially considered intermediaries in protein synthesis or degradation products of RNA metabolism, they are now understood as key regulatory molecules that influence genome organization and control gene expression [3].
Among non-coding RNAs (ncRNAs), microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) have garnered particular interest due to their relevant roles in a plethora of essential biological processes. miRNAs are small (~22 nucleotides), evolutionarily conserved molecules that primarily function in post-transcriptional gene silencing-related cell differentiation and proliferation, embryonic development, and homeostasis [4,5]. In contrast, lncRNAs are typically defined as transcripts longer than 200 nucleotides and are involved in a wide array of regulatory mechanisms, including chromatin remodeling, transcriptional control, and RNA processing [6]. The functional versatility of these ncRNAs underscores their significance in both normal physiology and disease [7].
miRNAs modulate gene expression by interacting with the 3′ untranslated regions (UTRs) of target mRNAs, usually suppressing translation or influencing stability and degradation [8,9]. Recent findings have revealed that many miRNAs map to exons of protein-coding genes (PCG), further increasing the complexity of these functional non-coding elements in the genomic regulatory organization [9]. In addition to modulating gene expression, miRNAs can also participate in intercellular communication, being secreted in extracellular vesicles [10] or serving as ligands to activate TLR signaling pathways, thereby inducing an immune response [11,12]. Numerous studies have demonstrated that dysregulated microRNA (miRNA) expression is intricately linked to the pathophysiology of a wide range of cellular processes. These include cardiovascular diseases [13], neurodegenerative disorders [14], immune system dysfunctions [15,16,17], and inflammatory conditions [18]. Notably, miRNA deregulation has been most extensively studied in the context of cancer, where it plays a critical role in tumor initiation, progression, and metastasis [19]. Additional evidence further supports the involvement of altered miRNA expression in other pathological states [18], highlighting their broad relevance in human disease. Since the early 2000s, accumulating evidence has underscored the role of dysregulated miRNAs, specifically functioning as oncogenes or tumor suppressors, in tumorigenesis [19]. A major seminal discovery in this field was the identification of the deletion of miR-5 and miR-6 in patients with chronic lymphocytic leukemia [18]. Another study investigated the let-7 family, which typically acts as a tumor suppressor by limiting tumor progression and metastasis [20]. Overall, miRNAs have emerged not only as valuable biomarkers for cancer development but also as important regulators of treatment responses, influencing drug resistance and therapeutic efficacy [21,22,23].
In recent decades, extensive research on various diseases, including cancer, diabetes, and neurological disorders [24], has also highlighted the functional importance and key roles of the microbiota, for example, maintaining homeostasis and modulating host gene expression [25]. Over the past few years, ncRNAs have also been identified as crucial regulators of the dynamic interplay between pathogenic microorganisms and their hosts [26]. Growing evidence suggest that microbial ncRNAs, such as miRNAs and lncRNAs, contribute to the alteration of host RNA expression profiles, either by producing their own ncRNAs to be incorporated by the host or by modulating host ncRNAs [27,28]. Several key processes in this context of host–microbiota interactions can be affected, including host inflammatory responses, cell invasion, tissue remodeling, and both innate and adaptive immunity [26,29,30]. In particular, viral miRNAs may contribute to regulating both pathogen and host gene expression, for example, to control the transition between viral phases, facilitate viral replication by enhancing cell survival, or influence immune responses. Thus, viral proteins and miRNAs could establish a favorable host environment for viral persistence [30]. For instance, miR-122 binding at the 5′ non-coding region of the hepatitis C virus is essential for viral RNA stability and increased infection rates [31,32]. In that sense, miR-122 binding has been reported to induce a sponge effect, sequestering free miR-122 at the site of infection. Consequently, the reduction in overall miR-122 levels potentially disrupts liver homeostasis and increases the risk of hepatocarcinoma [33]. In the same tumor context, Epstein–Barr virus (EBV) miRNAs have been observed to reduce the expression of the transcriptional repressor BCL6 in diffuse large B-cell lymphoma, which may be necessary to promote the survival of EBV-positive neoplastic cells [34]. Another strategy employed by viruses is the mimicry of host miRNAs. By replicating their sequence and structure, viral miRNAs can exploit the host gene regulatory machinery, controlling gene networks and facilitating disease progression and the establishment of chronic infections [35,36]. Viral miRNA mimicry typically retains only the similarity of the seed sequence, so that the set of potential target genes, known as the targetome, is not completely identical to that of the host miRNA [36]. For instance, the Kaposi’s sarcoma-associated herpesvirus (KSHV) encodes miR-K6-5p as a mimic of the tumor-suppressive cellular miR-15/16 miRNA family, so it can fine-tune its viral oncogene expression to prevent severe pathogenesis in a healthy host [37].
Similar to miRNAs, the regulatory function of lncRNAs in viral infection has been recognized for decades, with accumulating evidence demonstrating their involvement in various stages of the viral life cycle, resistance to antiviral immune response, and viral pathogenicity [38]. The PAN (polyadenylated nuclear RNA) lncRNA is implicated in multiple key stages of the viral life cycle, including the initiation of viral replication, repression of host immune regulator expression, regulation of cellular pluripotency, and mediation of host–pathogen interactions [38,39,40]. In addition to regulating gene expression, lncRNAs can directly interact with proteins, modulating viral processes and host–virus dynamics. For example, the lncRNA called low-molecular-weight tristeza 1 (LMT1) interacts with the Citrus Tristeza Virus (CTV) p33 protein, contributing to the maintenance of viral persistence throughout the life of the host organism [41]. Regarding cancer, EBV expresses lncRNAs that modulate host gene expression to facilitate infection and may contribute to tumor development. For instance, BART lncRNAs influence the regulation of genes associated with inflammatory responses, cell adhesion, and metastasis [42]. Similarly, it has been also demonstrated that the human cytomegalovirus lncRNA4.9 induces the rapid growth of triple-negative breast tumors in NOD/SCID gamma mice [43]. Beyond ncRNAs and miRNAs, there are other classes of viral non-coding RNA that enable viral entities to manipulate host cellular dynamics to facilitate their life cycle, such as the small nuclear ncRNAs (snRNAs) in γ-herpesviruses and the virus-associated (VA) ncRNAs in adenoviruses [44].
Due to the potential viral etiology of certain cancers, the analysis of miRNA profiles could be essential for identifying novel therapeutic targets [45]. Anal squamous cell carcinoma (ASCC) is one such cancer associated with viral presence. It is driven by infection with oncogenic human Alphapapillomavirus strains [46], and it accounts for up to 3% of gastrointestinal tumors [47]. In recent years, the global incidence of ASCC has increased, mainly among men who have sex with men and people living with HIV [48]. High-risk human papillomaviruses 16 (HPV) and 18 are responsible for the development of ASCC in approximately 80–85% of cases [49]. According to the grade of neoplasia, squamous intraepithelial lesions (SILs) typically precede the onset of ASCC and are categorized into low-grade SIL (LGSIL) and high-grade SIL (HGSIL) subtypes [50]. Infections with low-risk HPV subtypes, such as HPV6 and HPV11, are mainly linked to these SILs, including genital warts or condyloma acuminata [51,52]. Histology and immunohistochemistry remain the primary methods for diagnosing the SIL subtype, although there is still considerable variability in accurately defining the grade of the lesion [53,54]. Previous research has demonstrated that ASCC cases associated with HPV exhibit distinct clinicopathological covariates compared to those that are HPV-negative. Specifically, HPV-negative ASCCs are more often characterized by keratinization, lack of p16 immunoreactivity, and poorer survival rates compared to HPV-positive tumors [55,56]. A weakened immune system is another significant risk factor for ASCC, which is often found in individuals with HIV or organ transplant recipients who require long-term immunosuppressive therapy [57,58]. In addition, growing evidence suggests that microbial communities, including HPV, play a role in tumor development, particularly in HPV-associated malignancies [59,60]. Since preventing the progression of SILs could significantly reduce the risk of ASCC, identifying viral miRNAs in patient samples may provide valuable insights into early detection of the disease. Given the lack of reliable prognostic biomarkers, investigating transcriptomic alterations in the microbiome present in ASCC samples compared with SILs could be crucial for improving early diagnosis and risk stratification. The involvement of HPV16-encoded miRNAs, such as HPV16-miR-H1 and HPV16-miR-H6, in cervical cancer is well established, as they have been shown to enhance tumor progression by regulating enhancer activity and promoting cell migration [61]. However, little is known about the role of miRNAs in ASCC and their potential impact on tumor development.
Overall, although great efforts have been made to elucidate the functions of ncRNAs, the extent to which microbial organisms can regulate host gene expression through ncRNAs remains poorly understood. Considering the morphological and transcriptomic heterogeneity observed in different ASCC stages among patients and the established involvement of viruses in this malignancy, the detection of dysregulated viral miRNAs could open new avenues for improving clinical diagnosis and prognosis. By conforming to the FAIR data principles, in this study we leveraged already available total RNA datasets addressing different ASCC stages. Applying an up-to-date meta-transcriptomics workflow, our aim was to investigate and characterize the dysregulated viral miRNAs, together with their potential role in modulating host gene expression in a pathogenic context.

2. Results

2.1. Differential Microbial Abundance Across Precursor Lesions and ASCC

Our meta-transcriptomic pipeline enabled comprehensive profiling of all RNA species present in the samples analyzed by Lacunza et al. (2024) [62]. Out of these, ~1% were identified as non-human sequences and aligned to microorganisms’ genomes (see Methods). In total, 427 transcriptionally active microorganisms were identified in the patient samples. Among these, 88.60% belonged to bacteria, 5.00% to fungi, 2.14% to viruses, and 4.30% to archaea and protozoa. Next, we applied a novel computational approach to assess microbial differential abundance in four comparisons: (i) anal squamous cell carcinoma (ASCC) vs. high-grade squamous intraepithelial lesions (HGSILs), (ii) ASCC vs. low-grade squamous intraepithelial lesions (LGSILs), (iii) HGSILs vs. LGSILs, and (iv) a broader comparison of patients presenting ASCC vs. a grouping of high- and low-grade SILs (HGSILs + LGSILs; Supplementary Table S1).
When comparing ASCC vs. HGSILs, 85 differentially abundant microorganisms were detected, Alphapapillomavirus 10 (logFC = −7.05, FDR = 2.93 × 10−4) and Alphapapillomavirus 14 (logFC = −5.30, FDR = 1.31 × 10−3) exhibiting greater expression inhibition in ASCC. Between ASCC and LGSILs, 93 differentially abundant microorganisms were identified. In this comparison, Alphapapillomavirus 9 showed a higher abundance in ASCC samples (logFC = 5.91, FDR = 2.86 × 10−4), while Alphapapillomavirus 14 showed a lower abundance in ASCC (logFC = −4.09, FDR = 4.20 × 10−3). In addition, a significant decrease in the abundance of other Alphapapillomaviruses was observed, specifically Alphapapillomavirus 10 (logFC = −5.64, FDR = 9.35 × 10−4), i.e., a distinctive viral composition associated with ASCC. In the HGSIL vs. LGSIL comparison, we characterized 427 differentially abundant microorganisms (FDR < 0.05) corresponding to 20 microbial species (i.e., differentially abundant microorganisms), including bacteria and viruses. In this analysis, Alphapapillomavirus 9 was found to be significantly more abundant in the HGSIL samples against the LGSIL samples (logFC = 6.93, FDR = 1.68 × 10−4). Finally, we aimed to identify differences between patients with lesions and cancer. In the comparison between ASCC and LGSIL/HGSIL samples, we identified 47 differentially abundant microorganisms. Notably, Alphapapillomavirus 10 was significantly more abundant in LGSIL/HGSIL samples (logFC = –5.82, FDR = 1.20 × 10−3) (Figure 1; Supplementary Table S1).

2.2. Detection and Profiling of Viral miRNAs

Given the significant differential abundance of Alphapapillomavirus species 9, 10, and 14 observed in our analysis, we next investigated their potential impact on host transcriptomic regulation through viral miRNAs. These viral groups comprise multiple HPV genotypes associated with distinct pathological outcomes. Specifically, Alphapapillomavirus 9 includes high-risk genotypes such as HPV 16, 31, 33, 52, 58, and 67; Alphapapillomavirus 10 contains predominantly low-risk types, including HPV 6, 11, 13, 44, and 74; and Alphapapillomavirus 14 encompasses HPV 71, 90, and 106 [63]. This classification provided a framework for exploring the miRNA-mediated regulatory potential of individual strains within each viral group. Considering the limited information on miRNAs characterized in these HPVs of interest, we used their viral genomes to infer potential miRNAs (Supplementary Table S2). We predicted 90 significant viral miRNAs in this subset of Alphapapillomaviruses: 5 in HPV6, 32 in HPV11, 6 in HPV16, 1 in HPV33, 1 in HPV35, 15 in HPV39, 6 in HPV42, and 14 in HPV90. The sequences and details of the identified miRNAs are included in Supplementary Table S3.
To explore the potential regulatory roles of the newly identified miRNAs, we investigated their patterns of differential expression. For the comparisons between groups of patient samples depicted above revealed significant variations in viral miRNA expression only for HPV6, HPV11, and HPV90 (Figure 2). The highest number of differentially expressed miRNAs was observed in the comparisons of HGSIL vs. LGSIL (N = 36) and ASCC vs. HGSIL + LGSIL lesions (N = 37). In contrast, the comparisons of ASCC vs. HGSILs and ASCC vs. LGSILs resulted in a total of 27 and 26 differentially expressed miRNAs, respectively (Supplementary Table S4). In the case of HPV90, only the miRNA 3357-3373---AY057438.1 was differentially expressed, upregulated in the high-grade SIL samples for the HGSIL vs. LGSIL comparison (logFC = 1.85, FDR = 4.78 × 10−3) and downregulated in the cancerous samples for the ASCC vs. HGSIL comparison (logFC = −1.85, FDR = 0.03). Although Alphapapillomavirus 9 was the only virus found to be differentially abundant in the HGSIL vs. LGSIL comparison, no differentially expressed miRNAs associated with its constituent genotypes were detected. Consequently, this comparative was excluded from further analysis.

2.3. Prediction and Correlation of Viral miRNA Targets with Host Gene Expression

After identifying and characterizing the expression patterns of the viral miRNAs described above, we next investigated the differential transcriptional profiles of the human host that may be subject to regulation by these predicted viral miRNAs. To this end, we analyzed the human-derived fractions of the transcriptomic datasets. In the comparison among ASCC and HGSILs, 170 differentially expressed genes (FDR < 0.01) were identified, 43 downregulated and 127 upregulated (Figure 3A). Comparison between ASCC and LGSILs showed a higher number of affected genes, with 1927 differentially expressed genes, including 701 downregulated and 1226 upregulated genes (Figure 3B). In the last comparison grouping LGSILs and HGSILs into a single category versus ASCC, 1777 differentially expressed genes were detected, with 602 genes downregulated and 1175 upregulated (Figure 3C). These differentially expressed PCGs could be targets of the de novo miRNAs that we predicted above. Therefore, we next computed their potential interactions considering significant genes and HPV-miRNAs (FDR < 0.01 and FDR < 0.05, respectively; Supplementary Table S5).
The analysis of interactions between the identified miRNAs and differentially expressed PCGs yielded a variable number of potential targets across comparisons. In the ASCC vs. HGSIL comparison, 6407 potential target genes were identified, whereas in the ASCC vs. LGSIL comparison the number of targets reached 79,140 and in the comparison of ASCC vs. HGSILs + LGSILs a total of 95,036 targets were predicted. All identified interactions correspond to the 8-6mer type, a pairing pattern characterized by high affinity and specificity in miRNA binding to its target transcript (Supplementary Table S6).
In addition to the prediction of potential targets, an anti-correlation analysis was performed between the miRNAs and differentially expressed genes predicted to interact to investigate the potentially negative regulation of miRNAs on genes in the groups of interest (Figure 4). This approach allowed us to identify negative correlation profiles between miRNAs and their predicted target genes, which could be indicative of a post-transcriptional regulatory mechanism. To further validate and investigate this relationship in silico, we performed correlation analyses of the expression levels of the miRNAs and predicted target PCGs.
Notable differences were observed across the comparisons, reflecting varying degrees of miRNA–PCG regulatory interactions associated with lesion severity. The ASCC vs. HGSIL + LGSIL comparison showed the highest number of significant anti-correlations (177); in the ASCC vs. LGSIL comparison, 96 significant anti-correlations were identified; in contrast, the ASCC vs. HGSIL comparison revealed only 7 significant anti-correlations (Figure 4).
To examine in more detail the functional impact of these regulatory interactions, an enrichment analysis was performed on the differentially expressed PCGs potentially targeted by miRNAs. The comparison between ASCC and HGSILs potentially targeted by miRNAs identified 26 significant biological pathways, highlighting various key metabolic and cellular processes involved in the tumor progression (Figure 5A). These include several related to the cell cycle, such as meiotic nuclear division (p = 2.56 × 10−3) and the meiotic cell-cycle process (p = 3.06 × 10−3), indicating possible regulation of cell division in the early stages of the lesions. Additionally, several pathways associated with lipid metabolism were detected, such as regulation of the phosphatidylcholine metabolic process (p = 4.11 × 10−3), negative regulation of lipase activity (p = 6.56 × 10−3), and negative regulation of the fatty acid biosynthetic process (p = 6.56 × 10−3).
In the functional enrichment analysis of the differentially expressed miRNA-targeted PCGs in the comparison between ASCC and LGSILs, a total of 33 significant biological pathways were identified (Figure 5B). Among the most prominent pathways was the positive regulation of the Smoothened signaling pathway (p = 6.57 × 10−4), which is related to the Hedgehog signaling pathway, a key process in the regulation of cell development and proliferation. In addition, epithelial cell proliferation (p = 1.85 × 10−3) was the most enriched pathway, with eight distinct genes, closely followed by the regulation of the epithelial cell proliferation pathway (p = 1.19 × 10−3), which included six related genes. Other observed pathways, such as mesenchymal cell proliferation (p = 1.37 × 10−3) and the Wnt signaling pathway (p = 2.76 × 10−2), were identified during the enrichment analysis.
In the functional enrichment analysis of the differentially expressed miRNA-targeted PCGs in the comparison between ASCC and HGSILs + LGSILs, 46 significant biological pathways were identified (Figure 5C). Similar to the ASCC vs. LGSIL comparison, among the most prominent pathways were those associated with the cell cycle, such as the positive regulation of the meiotic cell cycle (p = 9.66 × 10−6) and meiotic nuclear division (p = 2.60 × 10−4), the Smoothened signaling pathway (p = 4.60 × 10−4), and mesenchymal cell proliferation (p = 7.50 × 10−4). Finally, other results include additional pathways related to viral processes (p = 2.55 × 10−3) and cell division (p = 1.95 × 10−2), highlighting potential mechanisms involved in disease progression (Supplementary Table S7).

3. Discussion

This study aimed to identify de novo viral miRNAs in previously published transcriptomic datasets derived from anal biopsy samples of patients with precursor lesions and ASCC [62]. To achieve this objective, a novel computational workflow was developed, and microbial abundance analysis was applied to the following sample group comparisons: (i) ASCC vs. HGSILs, (ii) ASCC vs. LGSILs, (iii) HGSILs vs. LGSILs, and (iv) ASCC vs. HGSILs + LGSILs. The number of differentially abundant microorganisms was higher in the ASCC vs. LGSIL comparison, while the lowest was observed in the HGSIL vs. LGSIL comparison. These results indicate a greater imbalance in microbial communities during carcinogenesis. Despite using a different methodology and experimental design to those of Lacunza et al., 2024 [62], our findings are consistent with their observations—notably, the high abundance of Alphapapillomaviruses 9, 10, and 14, which were among the most prevalent viral species identified across diagnostic groups in their study. These Alphapapillomavirus groups include different HPV strains with distinct carcinogenic potential. Alphapapillomavirus 9 is exclusively composed of high-risk HPV strains, including HPV-16, one of the most well-established oncogenic viruses [64]. In contrast, Alphapapillomaviruses 10 and 14 contain only low-risk HPV types, which are generally not associated with malignant progression. However, it is well described in the literature that numerous HPV types, including both high-risk and low-risk strains, have been implicated in carcinogenesis [65].
Since the discovery of the first viral miRNA in Epstein–Barr virus (EBV) in 2004, numerous others have been reported to play roles in various diseases, underscoring their potential involvement in viral pathogenesis and host regulation [61,66]. In this context, we aimed to investigate the role of viral miRNAs by characterizing the expression patterns of a newly predicted de novo miRNA dataset in order to assess their potential contribution to tumorigenesis during ASCC progression.
Previous studies have provided strong evidence that viral miRNAs, such as HPV16-miR-H1 and HPV16-miR-H6, alter the expression of host genes regulating the cell cycle and immune evasion in the pathogenesis of cervical cancer [61,67]. Other viral strains such as HPV6, HPV11, and HPV90 were reported in different cancer-related processes like genital warts and cervical intraepithelial lesions in women [68,69]. Although low-risk HPVs are generally considered less involved in carcinogenesis, some studies have identified cases where low-risk HPV presence may be also linked to anal cancer [70]. Additionally, coinfection with low-risk HPV strains, such as HPV6 and HPV18, alongside high-risk HPV types has been reported in anal cancer samples [71]. Building on this knowledge, our in silico analysis reveals that low-risk HPV viral miRNAs may also play a regulatory role in ASCC progression, a role that remains scarcely reported.
The high-confidence set of de novo HPV-miRNAs identified in this study is predicted to target host genes involved in critical tumorigenic pathways associated with the development and progression of ASCC, offering novel insights into host–pathogen regulatory interactions that remain incompletely understood. Supporting these findings, TargetScan predictions based on anti-correlated RNA expression revealed a substantial number of human genes potentially subject to negative regulation by viral miRNAs at more advanced stages of anal lesions. This observation aligns with the increased number of differentially expressed PCGs (FDR < 0.01) identified in comparisons involving ASCC, suggesting that miRNA-mediated disruptions of host transcriptional programs may contribute progressively to neoplastic transformation in ASCC.
Subsequently, in order to investigate the potential functions of these transcriptional profiles, functional enrichment analysis across the different comparisons was performed. When comparing ASCC vs. HGSILs, biological terms related to the cell cycle were identified, such as meiotic nuclear division and the meiotic cell-cycle process, indicating a possible regulation of cell division in the early stages of the lesions. Although the enrichment of meiosis-associated pathways in somatic cells was initially surprising, a closer examination of the genes driving this signal suggests that several may be aberrantly reactivated in cancer contexts—an aspect often overlooked or underrepresented in current Gene Ontology classifications. For instance, the gene SYCP2L, a paralog of the meiosis-specific synaptonemal complex protein 2 (SYCP2), which is involved in chromosome localization and axis assembly during meiotic recombination [72], was found to be upregulated in ASCC and annotated with the enriched GO terms. Given that SYCP2 has been reported as aberrantly expressed in ovarian and breast cancers—where its overexpression is associated with resistance to therapies targeting the DNA damage response [73]—it is plausible that SYCP2L may exhibit similar aberrant activation in the context of ASCC. While only limited studies have linked SYCP2L to female infertility [74,75], its sequence homology with SYCP2 raises the possibility of shared non-canonical functions, such as involvement in the negative regulation of cell death via DNA repair [73]. This potential role may be relevant in light of the enrichment of meiosis-related pathways observed in the ASCC vs. HGSIL comparison. This raises the possibility that the downregulation of HPV6-miRNA 839-861---NC_075235.1, identified as a high-confidence de novo miRNA in our analysis and putatively targeting SYCP2L expression, may function as an oncoMiR, promoting the progression from HGSIL to ASCC.
An additional functionally enriched term of interest was related to lipid metabolism, which aligns with the existing literature, highlighting the importance of lipid metabolic reprogramming in HPV-driven carcinogenesis. For instance, it has been shown that the expression of the HPV16 E6 oncoprotein upregulates miR-21, which in turn promotes the expression of several enzymes involved in lipid metabolism [76]. This misregulation of lipid metabolism allows HPV to modulate the composition of the cell membrane in order to control infection efficiency, evade the immune response, and manage energy reservoirs for cellular reprogramming in tumor contexts [77]. In our enrichment analysis, lipid metabolism alteration mostly corresponds to the overexpression of the APOC1 gene in ASCC, which has been also found to be upregulated in HPV-positive cervical cancer [78]. Among the findings derived from the de novo identified miRNAs in this study, we can highlight that the downregulation of HPV6-miRNA 3071-3094-+-NC_075235.1 seems to be highly associated with the upregulation of APOC1 in ASCC.
Additional relevant terms such as the enrichment related to interferon-alpha response pathways suggests a potential modulation of immune signaling during lesion progression. This observation aligns with previous studies reporting that HPV can affect interferon-mediated signaling, disrupt immune cell trafficking and adhesion, and alter cellular phenotypes. These disruptions are known to contribute to immune evasion, creating a microenvironment conducive to persistent infection and malignant transformation.
Similarly, the enrichment of pathways related to polyamine biosynthesis and amino compound metabolism highlights the importance of miRNA-targeted protein-coding genes involved in cell growth and proliferation. Prior research has shown that genes regulating polyamine metabolism are upregulated in HPV-infected cervical squamous cell carcinoma, where their activity may enhance the effects of polyamine oxidation. This interaction is proposed to inhibit apoptosis in keratinocytes carrying potentially oncogenic mutations, thereby promoting the survival and expansion of HPV-infected cells within a tumor-promoting context [76,79].
In the comparison between ASCC and LGSILs, enriched biological pathways associated with the positive regulation of mesenchymal cell proliferation were observed. This mesenchymal phenotype is known to be linked to increased invasiveness and metastasis [80]. It has been shown that the degradation of p-53 in HPV-infected epithelial cells in cervical cancer promotes epithelial–mesenchymal transition [81]. These findings suggest that the downregulated miRNAs in ASCC may potentiate this transition, contributing to enhancing the metastatic capacity of HPV-infected cells. Pathways related to the Wnt signaling pathway and cell polarity were also identified in our analysis, such as the non-canonical Wnt signaling pathway and the regulation of planar polarity, highlighting the importance of regulating cell behavior in terms of direction and spatial organization. The E6 and E7 oncoproteins, which are responsible for the oncogenesis of HPV, have been found to be involved in the regulation of the Wnt/β-catenin pathway [82]. Among the genes that are overexpressed in these pathways of ASCC vs. LGSIL analysis are GPC2 and GPC3 [83]. Both proteins have been implicated in the development of various types of tumors, such as hepatocellular carcinoma [84] and colorectal cancer [85]. Remarkably, we highlight the downregulation of the de novo HPV11-miRNA 4456-4476---NC_075235.1, which is predicted to target GPC3, suggesting a potential regulatory function in these processes. Taken together, these results suggest that the HPV-miRNAs identified as targeting genes involved in these functions are involved in a wide variety of biological and cellular processes, many of which are essential for development, cell proliferation, and tissue organization during injury progression.
Finally, the comparison between ASCC and LGSIL + HGSIL samples revealed the enrichment of pathways related to cell division and proliferation, including the regulation of the meiotic cell cycle and mesenchymal cell proliferation, highlighting key processes involved in tumor progression. As observed in the ASCC vs. HGSIL comparison, we obtained enriched meiosis-related pathways, whose genes involved in synaptonemal complex assembly have been associated with the regulation of genome integrity in cancer [86]. In our results, several enriched pathways related to the regulation of the oxidative stress response further highlight the potential involvement of miRNAs in protecting against cellular damage at the ASCC stage. Previous studies have shown that several antioxidant enzymes and detoxification pathways are also consistently linked to HPV-transformed cells, which appear to be well-adapted to highly oxidative environments [87]. Other important pathways include cytidine metabolism, the cytidine-to-uridine editing process, and homocysteine metabolism, reflecting the regulation of metabolic and epigenetic processes critical for genomic stability. It has been described that the APOBEC family of cytidine deaminases, which are involved in the enriched cytidine metabolism pathways identified in our analysis, may contribute to the generation of driver mutations within the helical domain of PIK3CA in HPV-positive cancers, thereby promoting HPV-mediated tumorigenesis [88,89]. Supporting this, our data showed the upregulation of APOBEC3B and APOBEC3F in ASCC, predicted to be regulated by the de novo HPV11-miRNAs 1892-1907-+-NC_075235.1 and 3071-3094-+-NC_075235.1, respectively. Lastly, we reported some pathways related to the viral life cycle and viral genome replication, such as negative regulation of viral genome replication, suggesting that HPV-miRNAs may also be involved in modulating the cell response to viral infection.
It is important to highlight that functional validation remains a critical next step to confirm the biological relevance of these miRNA–mRNA interactions. In this context, the work of Peronace et al. (2024) [90] provides a compelling example of in vitro validation of miRNA-based biomarkers as clinically useful tools. They highlighted the double methylation of FAM194 and has-miR-124-2 as a promising tool for diagnosing high-risk HPV-positive women with early-stage cervical lesions. Remarkably, their study reported a frequent association between HPV genotypes 16 and 18 and positive methylation of both genes, further supporting the relevance of these markers in HPV-driven carcinogenesis. Such findings underscore the importance of integrating molecular signatures into screening strategies for HPV-driven neoplasms, including ASCC. To advance beyond computational inference, future studies should incorporate experimental approaches widely used in the validation of viral miRNAs. For instance, the Dual-Luciferase® Reporter Assay has been successfully applied in hepatocellular carcinoma to confirm the regulation of PPM1A by HBV-miR-3 [91]. Additionally, overexpression or knockdown of specific viral miRNAs, combined with transcriptomic profiling (e.g., RNA-seq), could help uncover their broader effects on host gene expression. Techniques such as RNA immunoprecipitation (RIP) and CLIP-seq can identify direct physical interactions between miRNAs and their target transcripts within the RISC complex. High-throughput approaches like HITS-CLIP, PAR-CLIP, and qCLASH have also proven effective in mapping extensive viral miRNA–target networks, as demonstrated in herpesvirus models. The integration of these methodologies will be essential to validate and expand upon the regulatory mechanisms proposed here within the host–virus interaction landscape [36]. The integration of these techniques would contribute to robust validation of the postulated interactions and elucidate the functional impact of viral miRNAs on host gene regulation.
Beyond the biological findings, our study also serves as a proof of concept for the reanalysis of publicly available total RNA-Seq datasets to simultaneously profile host gene expression, viral content, and microbiota composition. This approach aligns with FAIR data principles and demonstrates the potential of leveraging existing sequencing data to uncover novel virus–host regulatory interactions. The pipeline we implemented is standardized and generalizable, offering a valuable resource for future investigations across other datasets, viral species, and HPV-associated diseases. As such, we believe this framework can contribute to advancing the understanding of virus-driven tumorigenesis through integrative, data-driven methodologies.

4. Material and Methods

4.1. Data Acquisition and Comparative Design

We used the transcriptomics dataset published by Lacunza et al., 2024 (GSE253560, GEO repository) [62]. This dataset comprises 31 LGSIL samples, 16 HGSIL samples, and 23 ASCC samples. ASCC samples correspond to patients with a confirmed cancer diagnosis, whereas LGSIL and HGSIL samples represent pre-invasive lesions of low- and high-grade severity, respectively. Details regarding sample collection, processing, and the RNA-Seq protocol are available in the original publication [62]. We focused our comparative analyses on specific pairwise contrasts, including ASCC versus LGSILs, ASCC versus HGSILs, LGSILs versus HGSILs, and ASCC versus LGSILs + HGSILs, in order to characterize more accurately the changes associated with the progression of pre-invasive lesions to invasive cancer.

4.2. Processing, Mapping, and Analysis of Human Transcriptomics Data

The reanalyzerGSE pipeline [92] was employed to conduct sample quality control, align sequences, and identify human differentially expressed genes. Raw reads were aligned using HISAT2 [93] against the PCGs of the Homo sapiens Gencode genome build GRCh38-v47 in very-sensitive mode. Once the reads were aligned, gene-level quantification was performed using featureCounts [94], with MOCB parameters applied: excluding multimapping reads, considering only properly paired reads aligned in a concordant manner, and including reads overlapping annotated features. Subsequently, gene expression matrices were generated for each sample, and differential expression analysis was carried out using the edgeR R package (v4.0.1) [95]. To account for differences in library size and sequencing depth across samples, counts were normalized using the trimmed mean of M-values (TMM) method [96], which is implemented by default in edgeR. This normalization ensures that observed differences in gene expression are not driven by technical biases. Multiple-comparisons adjustments were performed using the Benjamini–Hochberg method. The R package clusterProfiler [97] was used to conduct functional enrichment analysis and explore the biological implications of differentially expressed genes. Gene annotation was performed by mapping Entrez gene identifiers using an orgDB R object, generated through the AnnotationForge package, ensuring seamless integration with clusterProfiler for downstream analyses.
Unaligned reads were retained for taxonomic classification and microbial quantification by Kraken/Bracken [98]. The resulting reports were further analyzed to evaluate the microbial composition. To assess differences across conditions, we quantified the number of reads assigned to each taxon and processed them using the metagenomeSeq R package (v2.1.6) [99]. Taxa with a total read count of zero across all samples—typically introduced by the software despite absence—were removed prior to analysis. To correct for sequencing depth variation, read counts were normalized using the Cumulative Sum Scaling (CSS) method, which adjusts for library size while preserving the structure of sparse count data typical of microbiome datasets. This normalization approach mitigates the influence of highly abundant features and allows for more accurate comparisons across samples. Differential abundance analysis was then carried out using the zero-inflated Gaussian (ZIG) model implemented in metagenomeSeq [99].

4.3. Analysis Workflow for Viral miRNA Identification

To identify potential viral miRNAs with biological relevance in anal neoplasia, we employed a targeted two-step approach. First, we performed a differential abundance analysis of viral species across the comparisons performed among the ASCC, HGSIL, and LGSIL samples, identifying a subset of viruses whose presence varied significantly between lesion grades (see previous section). This step allowed us to prioritize a biologically relevant set of viral candidates that may be associated with tumor phenotype.
In the second step, we applied vsRNAfinder [100], a computational pipeline optimized for the detection and annotation of small RNAs (sRNAs), to identify putative viral miRNAs encoded by the genomes of the differentially abundant viruses. This tool requires as input raw RNA-Seq data in fastq format and the reference genome of each virus; alignments were performed using Bowtie v1.3.0 under default parameters [101]. Although the tool is capable of detecting various classes of small RNAs, our analysis focused exclusively on the identification of canonical and non-canonical viral miRNAs. For miRNA prediction, vsRNAfinder requires that the expression level within the coding region should be higher compared to its background. To account for this, the background region was defined as 10 nucleotides both upstream and downstream of the coding region. Additionally, a significance threshold of a p-value < 0.05 and a length range of 15–34 nucleotides were selected, based on the typical length of functional sRNAs, such as miRNAs and siRNAs, which generally range between 18 and 30 nt [101]. This threshold optimizes the accuracy of the analysis by minimizing background noise and focusing on the detection of true sRNAs, thereby reducing the likelihood of false positives.
As a validation method to ensure that the viral miRNAs detected by vsRNAfinder were not misclassified as human miRNAs, all identified sequences were queried against miRBase [102]. This analysis confirmed that none of the detected miRNAs exhibited full alignment with any known human miRNA, minimizing the likelihood of erroneous classification.
Additionally, to reduce the risk of false positives and ensure biological relevance, we implemented an additional filtering criterion: only miRNAs derived from viral species previously detected as significantly enriched (FDR ≤ 0.05) in the corresponding comparison groups were retained. While we acknowledge that viral miRNAs may still be expressed at low levels or in latent infections, our conservative strategy aimed to prioritize robust and phenotype-associated candidates for downstream analysis.

4.4. Prediction of miRNA-Regulated Target Genes

TargetScan (Release 8.0) was used with the list of differentially expressed PCGs to predict potential targets of the viral miRNAs [103]. To minimize the likelihood of false positives, only differentially expressed PCGs with an FDR < 0.01 were retained. Then, we used TargetScan to compare the 3′UTR sequences of differentially expressed genes against the seed sequences of our predicted miRNAs. The seed region, consisting of seven nucleotides spanning positions 2 to 8 of the miRNA sequence, is a critical determinant for miRNA-mRNA binding [104]. The predicted target genes can be categorized based on the degree of complementarity between the miRNA seed region and the 3′UTR sequence of the target genes. Depending on the number of aligned nucleotides, the predictions are classified as 6mer, 7mer, or 8mer matches. This classification offers insight into the binding affinity of a given miRNA to its target mRNA and, consequently, its potential regulatory effect on transcription. At this stage, all three types of binding sites were retained to avoid excluding any potential targets of interest.
Given that one of the primary mechanisms of miRNA-mediated regulation involves transcript degradation or translational repression, miRNAs typically display expression patterns that are inversely correlated with those of their target mRNAs. This negative correlation reflects the functional outcome of miRNA activity, whereby increased levels of an miRNA lead to reduced expression of its target genes. To prioritize biologically meaningful interactions and reduce the number of spurious associations, we limited our analysis to differentially expressed genes (DEGs) with a false discovery rate (FDR) < 0.01. Within this subset, we then assessed miRNA–mRNA expression relationships using the Spearman correlation coefficient, a non-parametric measure that captures monotonic associations without assuming linearity, in addition to applying a statistical significance threshold (p-value < 0.05).

5. Conclusions

In this study, we developed and implemented a novel in silico pipeline for the reanalysis of publicly available total RNA-seq data, enabling simultaneous profiling of host transcriptomics, microbial abundance, and viral miRNA discovery. This approach highlights the value of applying FAIR (Findable, Accessible, Interoperable, and Reusable) data principles to extract new layers of biological insight from existing datasets and demonstrates its potential applicability to other diseases and contexts.
Our integrative analysis provides a comprehensive characterization of HPV-derived miRNAs potentially involved in the progression of anal squamous cell carcinoma (ASCC). We identified novel viral miRNAs from HPV6, HPV11, and HPV90 with distinct expression patterns across lesion grades. These miRNAs appear to regulate host gene networks implicated in key tumorigenic processes, including cell-cycle control, epithelial–mesenchymal transition, lipid metabolism, immune modulation, and tissue organization.
Taken together, our findings suggest that viral miRNAs—particularly those encoded by low-risk HPV types—may contribute not only to the viral life cycle but also to the modulation of host gene expression and the promotion of neoplastic transformation. These results offer novel hypotheses about virus–host interactions in ASCC and lay the groundwork for future functional validation studies and biomarker discovery in HPV-associated cancers.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ncrna11030043/s1, Table S1: Differentially abundant microorganisms between comparisons. Table S2: HPV strains analyzed in the study, along with their corresponding chromosome IDs, reference genomes, and the presence or absence of detected miRNAs in the samples. Table S3: miRNA IDs that were predicted de novo. Table S4: Differentially expressed miRNAs between comparisons. Table S5: Differentially expressed genes between comparisons. Table S6: Predicted target genes of de novo miRNAs identified using TargetScan. Table S7: GO BP functional enrichment analysis of the differentially expressed genes that were predicted to be targets of de novo miRNAs.

Author Contributions

Conceptualization, L.C.T.-C., D.J.G. and E.A.-L.; methodology, L.C.T.-C., D.J.G., M.A.P.-B., E.A.-L. and J.L.R.; formal analysis, L.C.T.-C., D.J.G., M.A.P.-B. and J.L.R.; writing—original draft preparation, writing—review and editing, L.C.T.-C., D.J.G., M.A.P.-B., E.A.-L. and J.L.R. All authors have read and agreed to the published version of the manuscript.

Funding

L.C.T.-C. and M.A.P.-B. are supported by the Generation D initiative, promoted by Red.es, an organization attached to the Ministry for Digital Transformation and the Civil Service, for the attraction and retention of talent through grants and training contracts, financed by the Recovery, Transformation and Resilience Plan through the European Union’s Next Generation funds. J.L.R. is supported by a technical support staff grant from the Ministry of Science and Innovation of the Spanish Government (PTA2021-019864-I). D.J.G. is supported by a technical staff contract (E-18-2024-0094344), within the project BIOT22_00018_4, funded by the Spanish Ministry of Science, Innovation and Universities with funds from the European Union NextGenerationEU, from the Recovery, Transformation and Resilience Plan (PRTR-C17.I1) and from the Autonomous Community of Andalusia within the framework of the Biotechnology Plan Applied to Health”.

Data Availability Statement

The raw data analyzed in this study are available in the Gene Expression Omnibus (GEO) repository (https://www.ncbi.nlm.nih.gov/geo/), reference number GSE253560 (accessed on 21 October 2024). The data derived from the analyses are contained within the article and Supplementary Materials.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
Alpha-PV Alphapapillomavirus
ASCC Anal squamous cell carcinoma
HGSIL High-grade squamous intraepithelial lesion
HPV Human papillomavirus
LGSIL Low-grade squamous intraepithelial lesion
miRNAs MicroRNAs
ncRNAs Non-coding RNAs
SIL Squamous intraepithelial lesion
UTR Untranslated region

References

  1. Consortium, E.P. An integrated encyclopedia of DNA elements in the human genome. Nature 2012, 489, 57–74. [Google Scholar] [CrossRef] [PubMed]
  2. Palazzo, A.F.; Gregory, T.R. The case for junk DNA. PLoS Genet. 2014, 10, e1004351. [Google Scholar] [CrossRef]
  3. The expanding world of noncoding RNA biology. Nat. Cell Biol. 2022, 24, 1447.
  4. Gebert, L.F.R.; MacRae, I.J. Regulation of microRNA function in animals. Nat. Rev. Mol. Cell Biol. 2019, 20, 21–37. [Google Scholar] [CrossRef]
  5. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef]
  6. Huarte, M. The emerging role of lncRNAs in cancer. Nat. Med. 2015, 21, 1253–1261. [Google Scholar] [CrossRef]
  7. Nemeth, K.; Bayraktar, R.; Ferracin, M.; Calin, G.A. Non-coding RNAs in disease: From mechanisms to therapeutics. Nat. Rev. Genet. 2024, 25, 211–232. [Google Scholar] [CrossRef]
  8. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef]
  9. Gao, F.; Wang, F.; Chen, Y.; Deng, B.; Yang, F.; Cao, H.; Chen, J.; Chen, H.; Qi, F.; Kapranov, P. The human genome encodes a multitude of novel miRNAs. Nucleic Acids Res. 2025, 53, gkaf070. [Google Scholar] [CrossRef]
  10. Drula, R.; Pardini, B.; Fu, X.; De Los Santos, M.C.; Jurj, A.; Pang, L.; El-Daly, S.M.; Fabris, L.; Knutsen, E.; Dragomir, M.P.; et al. 17beta-estradiol promotes extracellular vesicle release and selective miRNA loading in ERalpha-positive breast cancer. Proc. Natl. Acad. Sci. USA 2023, 120, e2122053120. [Google Scholar] [CrossRef]
  11. Fabbri, M.; Paone, A.; Calore, F.; Galli, R.; Gaudio, E.; Santhanam, R.; Lovat, F.; Fadda, P.; Mao, C.; Nuovo, G.J.; et al. MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proc. Natl. Acad. Sci. USA 2012, 109, E2110–E2116. [Google Scholar] [CrossRef] [PubMed]
  12. Lehmann, S.M.; Kruger, C.; Park, B.; Derkow, K.; Rosenberger, K.; Baumgart, J.; Trimbuch, T.; Eom, G.; Hinz, M.; Kaul, D.; et al. An unconventional role for miRNA: Let-7 activates Toll-like receptor 7 and causes neurodegeneration. Nat. Neurosci. 2012, 15, 827–835. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, Y.; Chen, J.; Xiong, J.; Hu, J.Q.; Yang, L.Y.; Sun, Y.X.; Wei, Y.; Zhao, Y.; Li, X.; Zheng, Q.H.; et al. Potential cardiac-derived exosomal miRNAs involved in cardiac healing and remodeling after myocardial ischemia-reperfusion injury. Sci. Rep. 2024, 14, 24275. [Google Scholar] [CrossRef] [PubMed]
  14. Starhof, C.; Hejl, A.M.; Heegaard, N.H.H.; Carlsen, A.L.; Burton, M.; Lilje, B.; Winge, K. The biomarker potential of cell-free microRNA from cerebrospinal fluid in Parkinsonian Syndromes. Mov. Disord. 2019, 34, 246–254. [Google Scholar] [CrossRef]
  15. Negi, V.; Paul, D.; Das, S.; Bajpai, P.; Singh, S.; Mukhopadhyay, A.; Agrawal, A.; Ghosh, B. Altered expression and editing of miRNA-100 regulates iTreg differentiation. Nucleic Acids Res. 2015, 43, 8057–8065. [Google Scholar] [CrossRef] [PubMed]
  16. Alisi, L.; Giovannetti, F.; Armentano, M.; Lucchino, L.; Lambiase, A.; Bruscolini, A. Challenging corneal diseases and microRNA expression: Focus on rare diseases and new therapeutic frontiers. Surv. Ophthalmol. 2025, 70, 121–131. [Google Scholar] [CrossRef]
  17. Fehlmann, T.; Lehallier, B.; Schaum, N.; Hahn, O.; Kahraman, M.; Li, Y.; Grammes, N.; Geffers, L.; Backes, C.; Balling, R.; et al. Common diseases alter the physiological age-related blood microRNA profile. Nat. Commun. 2020, 11, 5958. [Google Scholar] [CrossRef]
  18. Calin, G.A.; Dumitru, C.D.; Shimizu, M.; Bichi, R.; Zupo, S.; Noch, E.; Aldler, H.; Rattan, S.; Keating, M.; Rai, K.; et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 2002, 99, 15524–15529. [Google Scholar] [CrossRef]
  19. Lin, S.; Gregory, R.I. MicroRNA biogenesis pathways in cancer. Nat. Rev. Cancer 2015, 15, 321–333. [Google Scholar] [CrossRef]
  20. Takamizawa, J.; Konishi, H.; Yanagisawa, K.; Tomida, S.; Osada, H.; Endoh, H.; Harano, T.; Yatabe, Y.; Nagino, M.; Nimura, Y.; et al. Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res. 2004, 64, 3753–3756. [Google Scholar] [CrossRef]
  21. Bhatnagar, N.; Li, X.; Padi, S.K.; Zhang, Q.; Tang, M.S.; Guo, B. Downregulation of miR-205 and miR-31 confers resistance to chemotherapy-induced apoptosis in prostate cancer cells. Cell Death Dis. 2010, 1, e105. [Google Scholar] [CrossRef] [PubMed]
  22. Xue, J.; Chi, Y.; Chen, Y.; Huang, S.; Ye, X.; Niu, J.; Wang, W.; Pfeffer, L.M.; Shao, Z.M.; Wu, Z.H.; et al. MiRNA-621 sensitizes breast cancer to chemotherapy by suppressing FBXO11 and enhancing p53 activity. Oncogene 2016, 35, 448–458. [Google Scholar] [CrossRef]
  23. Niu, J.; Xue, A.; Chi, Y.; Xue, J.; Wang, W.; Zhao, Z.; Fan, M.; Yang, C.H.; Shao, Z.M.; Pfeffer, L.M.; et al. Induction of miRNA-181a by genotoxic treatments promotes chemotherapeutic resistance and metastasis in breast cancer. Oncogene 2016, 35, 1302–1313. [Google Scholar] [CrossRef]
  24. Hou, K.; Wu, Z.X.; Chen, X.Y.; Wang, J.Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in health and diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef]
  25. Priya, S.; Burns, M.B.; Ward, T.; Mars, R.A.T.; Adamowicz, B.; Lock, E.F.; Kashyap, P.C.; Knights, D.; Blekhman, R. Identification of shared and disease-specific host gene-microbiome associations across human diseases using multi-omic integration. Nat. Microbiol. 2022, 7, 780–795. [Google Scholar] [CrossRef]
  26. Zhou, X.; Li, X.; Wu, M. miRNAs reshape immunity and inflammatory responses in bacterial infection. Signal Transduct. Target. Ther. 2018, 3, 14. [Google Scholar] [CrossRef]
  27. Liu, X.; Xiong, W.; Ye, M.; Lu, T.; Yuan, K.; Chang, S.; Han, Y.; Wang, Y.; Lu, L.; Bao, Y. Non-coding RNAs expression in SARS-CoV-2 infection: Pathogenesis, clinical significance, and therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 441. [Google Scholar] [CrossRef] [PubMed]
  28. Zhang, X.; Liang, Z.; Wang, C.; Shen, Z.; Sun, S.; Gong, C.; Hu, X. Viral Circular RNAs and Their Possible Roles in Virus-Host Interaction. Front. Immunol. 2022, 13, 939768. [Google Scholar] [CrossRef] [PubMed]
  29. Giza, D.E.; Fuentes-Mattei, E.; Bullock, M.D.; Tudor, S.; Goblirsch, M.J.; Fabbri, M.; Lupu, F.; Yeung, S.J.; Vasilescu, C.; Calin, G.A. Cellular and viral microRNAs in sepsis: Mechanisms of action and clinical applications. Cell Death Differ. 2016, 23, 1906–1918. [Google Scholar] [CrossRef]
  30. Abbas, M.N.; Kausar, S.; Asma, B.; Ran, W.; Li, J.; Lin, Z.; Li, T.; Cui, H. MicroRNAs reshape the immunity of insects in response to bacterial infection. Front. Immunol. 2023, 14, 1176966. [Google Scholar] [CrossRef]
  31. Jopling, C.L.; Yi, M.; Lancaster, A.M.; Lemon, S.M.; Sarnow, P. Modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA. Science 2005, 309, 1577–1581. [Google Scholar] [CrossRef] [PubMed]
  32. Thibault, P.A.; Huys, A.; Amador-Canizares, Y.; Gailius, J.E.; Pinel, D.E.; Wilson, J.A. Regulation of Hepatitis C Virus Genome Replication by Xrn1 and MicroRNA-122 Binding to Individual Sites in the 5’ Untranslated Region. J. Virol. 2015, 89, 6294–6311. [Google Scholar] [CrossRef]
  33. Luna, J.M.; Scheel, T.K.; Danino, T.; Shaw, K.S.; Mele, A.; Fak, J.J.; Nishiuchi, E.; Takacs, C.N.; Catanese, M.T.; de Jong, Y.P.; et al. Hepatitis C virus RNA functionally sequesters miR-122. Cell 2015, 160, 1099–1110. [Google Scholar] [CrossRef]
  34. Martin-Perez, D.; Vargiu, P.; Montes-Moreno, S.; Leon, E.A.; Rodriguez-Pinilla, S.M.; Lisio, L.D.; Martinez, N.; Rodriguez, R.; Mollejo, M.; Castellvi, J.; et al. Epstein-Barr virus microRNAs repress BCL6 expression in diffuse large B-cell lymphoma. Leukemia 2012, 26, 180–183. [Google Scholar] [CrossRef]
  35. Skalsky, R.L.; Cullen, B.R. Viruses, microRNAs, and host interactions. Annu. Rev. Microbiol. 2010, 64, 123–141. [Google Scholar] [CrossRef] [PubMed]
  36. Diggins, N.L.; Hancock, M.H. Viral miRNA regulation of host gene expression. Semin. Cell Dev. Biol. 2023, 146, 2–19. [Google Scholar] [CrossRef]
  37. Morrison, K.; Manzano, M.; Chung, K.; Schipma, M.J.; Bartom, E.T.; Gottwein, E. The Oncogenic Kaposi’s Sarcoma-Associated Herpesvirus Encodes a Mimic of the Tumor-Suppressive miR-15/16 miRNA Family. Cell Rep. 2019, 29, 2961–2969.e6. [Google Scholar] [CrossRef] [PubMed]
  38. Li, Z.; Gao, J.; Xiang, X.; Deng, J.; Gao, D.; Sheng, X. Viral long non-coding RNA regulates virus life-cycle and pathogenicity. Mol. Biol. Rep. 2022, 49, 6693–6700. [Google Scholar] [CrossRef]
  39. Borah, S.; Darricarrere, N.; Darnell, A.; Myoung, J.; Steitz, J.A. A viral nuclear noncoding RNA binds re-localized poly(A) binding protein and is required for late KSHV gene expression. PLoS Pathog. 2011, 7, e1002300. [Google Scholar] [CrossRef]
  40. Rossetto, C.C.; Pari, G.S. Kaposi’s sarcoma-associated herpesvirus noncoding polyadenylated nuclear RNA interacts with virus- and host cell-encoded proteins and suppresses expression of genes involved in immune modulation. J. Virol. 2011, 85, 13290–13297. [Google Scholar] [CrossRef]
  41. Kang, S.H.; Aknadibossian, V.; Kharel, L.; Mudiyanselage, S.D.D.; Wang, Y.; Folimonova, S.Y. The Intriguing Conundrum of a Nonconserved Multifunctional Protein of Citrus Tristeza Virus That Interacts with a Viral Long Non-Coding RNA. Viruses 2021, 13, 2129. [Google Scholar] [CrossRef] [PubMed]
  42. Marquitz, A.R.; Mathur, A.; Edwards, R.H.; Raab-Traub, N. Host Gene Expression Is Regulated by Two Types of Noncoding RNAs Transcribed from the Epstein-Barr Virus BamHI A Rightward Transcript Region. J. Virol. 2015, 89, 11256–11268. [Google Scholar] [CrossRef] [PubMed]
  43. Kumar, A.; Tripathy, M.K.; Pasquereau, S.; Al Moussawi, F.; Abbas, W.; Coquard, L.; Khan, K.A.; Russo, L.; Algros, M.P.; Valmary-Degano, S.; et al. The Human Cytomegalovirus Strain DB Activates Oncogenic Pathways in Mammary Epithelial Cells. EBioMedicine 2018, 30, 167–183. [Google Scholar] [CrossRef]
  44. Gorbea, C.; Mosbruger, T.; Cazalla, D. A viral Sm-class RNA base-pairs with mRNAs and recruits microRNAs to inhibit apoptosis. Nature 2017, 550, 275–279. [Google Scholar] [CrossRef]
  45. Marima, R.; Hull, R.; Mbeje, M.; Molefi, T.; Mathabe, K.; Elbagory, A.M.; Demetriou, D.; Dlamini, Z. Role of Precision Oncology in Type II Endometrial and Prostate Cancers in the African Population: Global Cancer Genomics Disparities. Int. J. Mol. Sci. 2022, 23, 628. [Google Scholar] [CrossRef] [PubMed]
  46. Hoots, B.E.; Palefsky, J.M.; Pimenta, J.M.; Smith, J.S. Human papillomavirus type distribution in anal cancer and anal intraepithelial lesions. Int. J. Cancer 2009, 124, 2375–2383. [Google Scholar] [CrossRef]
  47. Islami, F.; Ferlay, J.; Lortet-Tieulent, J.; Bray, F.; Jemal, A. International trends in anal cancer incidence rates. Int. J. Epidemiol. 2017, 46, 924–938. [Google Scholar] [CrossRef]
  48. Clifford, G.M.; Georges, D.; Shiels, M.S.; Engels, E.A.; Albuquerque, A.; Poynten, I.M.; de Pokomandy, A.; Easson, A.M.; Stier, E.A. A meta-analysis of anal cancer incidence by risk group: Toward a unified anal cancer risk scale. Int. J. Cancer 2021, 148, 38–47. [Google Scholar] [CrossRef]
  49. Daling, J.R.; Madeleine, M.M.; Johnson, L.G.; Schwartz, S.M.; Shera, K.A.; Wurscher, M.A.; Carter, J.J.; Porter, P.L.; Galloway, D.A.; McDougall, J.K. Human papillomavirus, smoking, and sexual practices in the etiology of anal cancer. Cancer 2004, 101, 270–280. [Google Scholar] [CrossRef]
  50. Pineda, C.E.; Welton, M.L. Management of anal squamous intraepithelial lesions. Clin. Colon Rectal Surg. 2009, 22, 94–101. [Google Scholar] [CrossRef]
  51. Cornall, A.M.; Roberts, J.M.; Garland, S.M.; Hillman, R.J.; Grulich, A.E.; Tabrizi, S.N. Anal and perianal squamous carcinomas and high-grade intraepithelial lesions exclusively associated with “low-risk” HPV genotypes 6 and 11. Int. J. Cancer 2013, 133, 2253–2258. [Google Scholar] [CrossRef]
  52. Liu, M.Z.; Hung, Y.P.; Huang, E.C.; Howitt, B.E.; Nucci, M.R.; Crum, C.P. HPV 6-associated HSIL/Squamous Carcinoma in the Anogenital Tract. Int. J. Gynecol. Pathol. 2019, 38, 493–497. [Google Scholar] [CrossRef]
  53. Vyas, M.; Gonzalez, R.S. Anal intraepithelial neoplasia: A review of terminology, differential diagnoses, and patient management. Hum. Pathol. 2023, 132, 56–64. [Google Scholar] [CrossRef]
  54. Madahian, S.; Judelson, R.; Zhu, X.; Meng, X.; Dresser, K.; Hutchinson, L.; Bledsoe, J.R. CD56 expression in basaloid anal squamous cell carcinoma—A potential diagnostic pitfall. Ann. Diagn. Pathol. 2021, 53, 151758. [Google Scholar] [CrossRef]
  55. Zhu, X.; Jamshed, S.; Zou, J.; Azar, A.; Meng, X.; Bathini, V.; Dresser, K.; Strock, C.; Yalamarti, B.; Yang, M.; et al. Molecular and immunophenotypic characterization of anal squamous cell carcinoma reveals distinct clinicopathologic groups associated with HPV and TP53 mutation status. Mod. Pathol. 2021, 34, 1017–1030. [Google Scholar] [CrossRef]
  56. Sun, G.; Dong, X.; Tang, X.; Qu, H.; Zhang, H.; Zhao, E. The prognostic value of HPV combined p16 status in patients with anal squamous cell carcinoma: A meta-analysis. Oncotarget 2018, 9, 8081–8088. [Google Scholar] [CrossRef]
  57. Silverberg, M.J.; Lau, B.; Justice, A.C.; Engels, E.; Gill, M.J.; Goedert, J.J.; Kirk, G.D.; D’Souza, G.; Bosch, R.J.; Brooks, J.T.; et al. Risk of anal cancer in HIV-infected and HIV-uninfected individuals in North America. Clin. Infect. Dis. 2012, 54, 1026–1034. [Google Scholar] [CrossRef]
  58. Sunesen, K.G.; Norgaard, M.; Thorlacius-Ussing, O.; Laurberg, S. Immunosuppressive disorders and risk of anal squamous cell carcinoma: A nationwide cohort study in Denmark, 1978-2005. Int. J. Cancer 2010, 127, 675–684. [Google Scholar] [CrossRef]
  59. Ron, R.; Cabello, A.; Gosalbes, M.J.; Sanchez-Conde, M.; Talavera-Rodriguez, A.; Zamora, J.; Monge-Maillo, B.; Jimenez, D.; Martinez-Sanz, J.; Lopez, Y.; et al. Exploiting the Microbiota for the Diagnosis of Anal Precancerous Lesions in Men Who Have Sex With Men. J. Infect. Dis. 2021, 224, 1247–1256. [Google Scholar] [CrossRef]
  60. Serrano-Villar, S.; Tincati, C.; Raju, S.C.; Saenz, J.S.; Moreno, E.; Bargiela, R.; Cabello-Ubeda, A.; Sendagorta, E.; Kurz, A.; Perez Molina, J.A.; et al. Microbiome-derived cobalamin and succinyl-CoA as biomarkers for improved screening of anal cancer. Nat. Med. 2023, 29, 1738–1749. [Google Scholar] [CrossRef]
  61. Wang, Y.; Wang, X.; Liu, Y.; He, Y.; Duan, X.; Li, Q.; Huang, Y.; Xu, G.; Lu, Q. HPV16-miRNAs exert oncogenic effects through enhancers in human cervical cancer. Cancer Cell Int. 2024, 24, 172. [Google Scholar] [CrossRef]
  62. Lacunza, E.; Fink, V.; Salas, M.E.; Gun, A.M.; Basiletti, J.A.; Picconi, M.A.; Golubicki, M.; Robbio, J.; Kujaruk, M.; Iseas, S.; et al. Transcriptome and microbiome-immune changes across preinvasive and invasive anal cancer lesions. JCI Insight 2024, 9, e180907. [Google Scholar] [CrossRef]
  63. Schoch, C.L.; Ciufo, S.; Domrachev, M.; Hotton, C.L.; Kannan, S.; Khovanskaya, R.; Leipe, D.; McVeigh, R.; O’Neill, K.; Robbertse, B.; et al. NCBI Taxonomy: A comprehensive update on curation, resources and tools. Database 2020, 2020, baaa062. [Google Scholar] [CrossRef]
  64. Zhang, Y.; Qiu, K.; Ren, J.; Zhao, Y.; Cheng, P. Roles of human papillomavirus in cancers: Oncogenic mechanisms and clinical use. Signal Transduct. Target. Ther. 2025, 10, 44. [Google Scholar] [CrossRef]
  65. Burd, E.M. Human papillomavirus and cervical cancer. Clin. Microbiol. Rev. 2003, 16, 1–17. [Google Scholar] [CrossRef]
  66. Grundhoff, A.; Sullivan, C.S. Virus-encoded microRNAs. Virology 2011, 411, 325–343. [Google Scholar] [CrossRef]
  67. Hull, R.; Marima, R.; Alaouna, M.; Demetriou, D.; Reis, R.M.; Molefi, T.; Dlamini, Z. Viral Encoded miRNAs in Tumorigenesis: Theranostic Opportunities in Precision Oncology. Microorganisms 2022, 10, 1448. [Google Scholar] [CrossRef]
  68. Quiroga-Garza, G.; Zhou, H.; Mody, D.R.; Schwartz, M.R.; Ge, Y. Unexpected high prevalence of HPV 90 infection in an underserved population: Is it really a low-risk genotype? Arch. Pathol. Lab. Med. 2013, 137, 1569–1573. [Google Scholar] [CrossRef]
  69. Liang, L.A.; Tanaka, L.F.; Radde, K.; Bussas, U.; Ikenberg, H.; Heideman, D.A.M.; Meijer, C.; Blettner, M.; Klug, S.J. Population-based age- and type-specific prevalence of human papillomavirus among non-vaccinated women aged 30 years and above in Germany. BMC Infect. Dis. 2024, 24, 1008. [Google Scholar] [CrossRef]
  70. Silva, L.L.D.; Teles, A.M.; Santos, J.M.O.; Souza de Andrade, M.; Medeiros, R.; Faustino-Rocha, A.I.; Oliveira, P.A.; Dos Santos, A.P.A.; Ferreira Lopes, F.; Braz, G.; et al. Malignancy Associated with Low-Risk HPV6 and HPV11: A Systematic Review and Implications for Cancer Prevention. Cancers 2023, 15, 4068. [Google Scholar] [CrossRef]
  71. Garbuglia, A.R.; Gentile, M.; Del Nonno, F.; Lorenzini, P.; Lapa, D.; Lupi, F.; Pinnetti, C.; Baiocchini, A.; Libertone, R.; Cicalini, S.; et al. An anal cancer screening program for MSM in Italy: Prevalence of multiple HPV types and vaccine-targeted infections. J. Clin. Virol. 2015, 72, 49–54. [Google Scholar] [CrossRef]
  72. West, A.M.; Rosenberg, S.C.; Ur, S.N.; Lehmer, M.K.; Ye, Q.; Hagemann, G.; Caballero, I.; Uson, I.; MacQueen, A.J.; Herzog, F.; et al. A conserved filamentous assembly underlies the structure of the meiotic chromosome axis. Elife 2019, 8, e40372. [Google Scholar] [CrossRef]
  73. Wang, Y.; Gao, B.; Zhang, L.; Wang, X.; Zhu, X.; Yang, H.; Zhang, F.; Zhu, X.; Zhou, B.; Yao, S.; et al. Meiotic protein SYCP2 confers resistance to DNA-damaging agents through R-loop-mediated DNA repair. Nat. Commun. 2024, 15, 1568. [Google Scholar] [CrossRef]
  74. Zhou, J.; Stein, P.; Leu, N.A.; Chmatal, L.; Xue, J.; Ma, J.; Huang, X.; Lampson, M.A.; Schultz, R.M.; Wang, P.J. Accelerated reproductive aging in females lacking a novel centromere protein SYCP2L. Hum. Mol. Genet. 2015, 24, 6505–6514. [Google Scholar] [CrossRef]
  75. He, W.B.; Tan, C.; Zhang, Y.X.; Meng, L.L.; Gong, F.; Lu, G.X.; Lin, G.; Du, J.; Tan, Y.Q. Homozygous variants in SYCP2L cause premature ovarian insufficiency. J. Med. Genet. 2021, 58, 168–172. [Google Scholar] [CrossRef]
  76. Ben, W.; Yang, Y.; Yuan, J.; Sun, J.; Huang, M.; Zhang, D.; Zheng, J. Human papillomavirus 16 E6 modulates the expression of host microRNAs in cervical cancer. Taiwan. J. Obstet. Gynecol. 2015, 54, 364–370. [Google Scholar] [CrossRef]
  77. Shakib, K.; Norman, J.T.; Fine, L.G.; Brown, L.R.; Godovac-Zimmermann, J. Proteomics profiling of nuclear proteins for kidney fibroblasts suggests hypoxia, meiosis, and cancer may meet in the nucleus. Proteomics 2005, 5, 2819–2838. [Google Scholar] [CrossRef]
  78. Zhang, Y.; Zhang, Y.; Pan, C.; Wang, W.; Yu, Y. HPV-driven heterogeneity in cervical cancer: Study on the role of epithelial cells and myofibroblasts in the tumor progression based on single-cell RNA sequencing analysis. PeerJ 2024, 12, e18158. [Google Scholar] [CrossRef]
  79. Nikmanesh, N.; Safarpour, A.R.; Niknam, R.; Nikmanesh, Y. Short Communication on HPV Infection, Pathogenesis of Cancer, Immune Cells Involved in Infection, Vaccination, and Suggested Treatments. J. Cancer Immunol. 2025, 7, 72–80. [Google Scholar] [CrossRef]
  80. Yeung, K.T.; Yang, J. Epithelial-mesenchymal transition in tumor metastasis. Mol. Oncol. 2017, 11, 28–39. [Google Scholar] [CrossRef]
  81. Han, Y.; Li, Q.; Ling, C.; Jin, M.; Li, D.; Zhong, J.; Wang, L. HPV-Induced MiR-21 Promotes Epithelial Mesenchymal Transformation and Tumor Progression in Cervical Cancer Cells through the TGFbeta R2/hTERC Pathway. Contrast Media Mol. Imaging 2022, 2022, 6297694. [Google Scholar] [CrossRef] [PubMed]
  82. Bello, J.O.; Nieva, L.O.; Paredes, A.C.; Gonzalez, A.M.; Zavaleta, L.R.; Lizano, M. Regulation of the Wnt/beta-Catenin Signaling Pathway by Human Papillomavirus E6 and E7 Oncoproteins. Viruses 2015, 7, 4734–4755. [Google Scholar] [CrossRef]
  83. Kolluri, A.; Ho, M. The Role of Glypican-3 in Regulating Wnt, YAP, and Hedgehog in Liver Cancer. Front. Oncol. 2019, 9, 708. [Google Scholar] [CrossRef]
  84. Sun, B.; Huang, Z.; Wang, B.; Yu, Y.; Lin, S.; Luo, L.; Wang, Y.; Huang, Z. Significance of Glypican-3 (GPC3) Expression in Hepatocellular Cancer Diagnosis. Med. Sci. Monit. 2017, 23, 850–855. [Google Scholar] [CrossRef]
  85. Wang, R.; Lin, X. GPC2 as a diagnostic and prognostic marker regulated progression of colorectal cancer. Arab. J. Gastroenterol. 2024, 25, 51–57. [Google Scholar] [CrossRef]
  86. Hosoya, N.; Miyagawa, K. Synaptonemal complex proteins modulate the level of genome integrity in cancers. Cancer Sci. 2021, 112, 989–996. [Google Scholar] [CrossRef] [PubMed]
  87. De Marco, F. Oxidative stress and HPV carcinogenesis. Viruses 2013, 5, 708–731. [Google Scholar] [CrossRef]
  88. Henderson, S.; Chakravarthy, A.; Su, X.; Boshoff, C.; Fenton, T.R. APOBEC-mediated cytosine deamination links PIK3CA helical domain mutations to human papillomavirus-driven tumor development. Cell Rep. 2014, 7, 1833–1841. [Google Scholar] [CrossRef] [PubMed]
  89. Warren, C.J.; Westrich, J.A.; Doorslaer, K.V.; Pyeon, D. Roles of APOBEC3A and APOBEC3B in Human Papillomavirus Infection and Disease Progression. Viruses 2017, 9, 233. [Google Scholar] [CrossRef]
  90. Peronace, C.; Cione, E.; Abrego-Guandique, D.M.; Fazio, M.; Panduri, G.; Caroleo, M.C.; Cannataro, R.; Minchella, P. FAM19A4 and hsa-miR124-2 Double Methylation as Screening for ASC-H- and CIN1 HPV-Positive Women. Pathogens 2024, 13, 312. [Google Scholar] [CrossRef]
  91. Chen, H.; Cao, D.; Han, N.; Zhang, M.; Jiang, W.; Wang, X.; Zeng, Q.; Tang, H. Hepatitis B Virus-Encoded MicroRNA (HBV-miR-3) Inhibits FIH-1 Expression to Promote Tumor Angiogenesis in HBV-Related Hepatocellular Carcinoma. J. Hepatocell. Carcinoma 2023, 10, 2337–2353. [Google Scholar] [CrossRef] [PubMed]
  92. Ruiz, J.L.; Terrón-Camero, L.C.; Castillo-González, J.; Fernández-Rengel, I.; Delgado, M.; Gonzalez-Rey, E.; Andrés-León, E. reanalyzerGSE: Tackling the everlasting lack of reproducibility and reanalyses in transcriptomics. bioRxiv 2023. bioRxiv:12.548663. [Google Scholar] [CrossRef]
  93. Kim, D.; Paggi, J.M.; Park, C.; Bennett, C.; Salzberg, S.L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 2019, 37, 907–915. [Google Scholar] [CrossRef]
  94. Liao, Y.; Smyth, G.K.; Shi, W. featureCounts: An efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 2014, 30, 923–930. [Google Scholar] [CrossRef]
  95. Nikolayeva, O.; Robinson, M.D. edgeR for differential RNA-seq and ChIP-seq analysis: An application to stem cell biology. Methods Mol. Biol. 2014, 1150, 45–79. [Google Scholar]
  96. Robinson, M.D.; Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 2010, 11, R25. [Google Scholar] [CrossRef] [PubMed]
  97. Yu, G.; Wang, L.G.; Han, Y.; He, Q.Y. clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS J. Integr. Biol. 2012, 16, 284–287. [Google Scholar] [CrossRef]
  98. Wood, D.E.; Salzberg, S.L. Kraken: Ultrafast metagenomic sequence classification using exact alignments. Genome Biol. 2014, 15, R46. [Google Scholar] [CrossRef]
  99. Paulson, J.N.; Stine, O.C.; Bravo, H.C.; Pop, M. Differential abundance analysis for microbial marker-gene surveys. Nat. Methods 2013, 10, 1200–1202. [Google Scholar] [CrossRef]
  100. Cai, Z.; Fu, P.; Qiu, Y.; Wu, A.; Zhang, G.; Wang, Y.; Jiang, T.; Ge, X.Y.; Zhu, H.; Peng, Y. vsRNAfinder: A novel method for identifying high-confidence viral small RNAs from small RNA-Seq data. Brief. Bioinform. 2022, 23, bbac496. [Google Scholar] [CrossRef]
  101. Langmead, B.; Trapnell, C.; Pop, M.; Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009, 10, R25. [Google Scholar] [CrossRef] [PubMed]
  102. Kozomara, A.; Birgaoanu, M.; Griffiths-Jones, S. miRBase: From microRNA sequences to function. Nucleic Acids Res. 2019, 47, D155–D162. [Google Scholar] [CrossRef] [PubMed]
  103. McGeary, S.E.; Lin, K.S.; Shi, C.Y.; Pham, T.M.; Bisaria, N.; Kelley, G.M.; Bartel, D.P. The biochemical basis of microRNA targeting efficacy. Science 2019, 366, eaav1741. [Google Scholar] [CrossRef] [PubMed]
  104. Chipman, L.B.; Pasquinelli, A.E. miRNA Targeting: Growing beyond the Seed. Trends Genet. 2019, 35, 215–222. [Google Scholar] [CrossRef]
Figure 1. Differential abundance analysis of viruses across the four comparisons: anal squamous cell carcinoma (ASCC) vs. high-grade squamous intraepithelial lesions (HGSILs), ASCC vs. low-grade squamous intraepithelial lesions (LGSILs), HGSILs vs. LGSILs, and ASCC vs. LGSILs and HGSILs. Circles represent downregulated viruses, while hexagons represent upregulated viruses among the samples. The size of each point corresponds to the logFC (fold change) value. False discovery rate (FDR) is represented using a color gradient.
Figure 1. Differential abundance analysis of viruses across the four comparisons: anal squamous cell carcinoma (ASCC) vs. high-grade squamous intraepithelial lesions (HGSILs), ASCC vs. low-grade squamous intraepithelial lesions (LGSILs), HGSILs vs. LGSILs, and ASCC vs. LGSILs and HGSILs. Circles represent downregulated viruses, while hexagons represent upregulated viruses among the samples. The size of each point corresponds to the logFC (fold change) value. False discovery rate (FDR) is represented using a color gradient.
Ncrna 11 00043 g001
Figure 2. Volcano plots showing differentially expressed HPV miRNAs across the following comparisons: (A) high-grade squamous intraepithelial lesions (HGSILs) vs. low-grade squamous intraepithelial lesions (LGSILs), (B) anal squamous cell carcinoma (ASCC) vs. HGSILs, (C) ASCC vs. LGSILs, and (D) ASCC vs. combined HGSILs + LGSILs. Colored dots indicate significant miRNAs (FDR < 0.05), yellow dots represent upregulated miRNAs, and blue dots indicate downregulated ones. For each comparison, the most significantly upregulated or downregulated miRNAs were listed according to their fold changes (|logFC|).
Figure 2. Volcano plots showing differentially expressed HPV miRNAs across the following comparisons: (A) high-grade squamous intraepithelial lesions (HGSILs) vs. low-grade squamous intraepithelial lesions (LGSILs), (B) anal squamous cell carcinoma (ASCC) vs. HGSILs, (C) ASCC vs. LGSILs, and (D) ASCC vs. combined HGSILs + LGSILs. Colored dots indicate significant miRNAs (FDR < 0.05), yellow dots represent upregulated miRNAs, and blue dots indicate downregulated ones. For each comparison, the most significantly upregulated or downregulated miRNAs were listed according to their fold changes (|logFC|).
Ncrna 11 00043 g002
Figure 3. Volcano plots represent differentially expressed protein-coding genes (PCGs) in three comparisons: (A) anal squamous cell carcinoma (ASCC) vs. high-grade squamous intraepithelial lesions (HGSILs), (B) HGSILs vs. low-grade squamous intraepithelial lesions (LGSILs), and (C) ASCC vs. combined HGSILs + LGSILs. Colored dots represent statistically significant miRNAs (FDR < 0.05): yellow points represent upregulated PCGs, while blue points depict downregulated PCGs. Labeled genes are those with the largest absolute logFC (fold change) values and statistical significance. For each comparison, the most significantly upregulated or downregulated genes were listed according to their fold changes (|logFC|).
Figure 3. Volcano plots represent differentially expressed protein-coding genes (PCGs) in three comparisons: (A) anal squamous cell carcinoma (ASCC) vs. high-grade squamous intraepithelial lesions (HGSILs), (B) HGSILs vs. low-grade squamous intraepithelial lesions (LGSILs), and (C) ASCC vs. combined HGSILs + LGSILs. Colored dots represent statistically significant miRNAs (FDR < 0.05): yellow points represent upregulated PCGs, while blue points depict downregulated PCGs. Labeled genes are those with the largest absolute logFC (fold change) values and statistical significance. For each comparison, the most significantly upregulated or downregulated genes were listed according to their fold changes (|logFC|).
Ncrna 11 00043 g003
Figure 4. Networks representing the correlation patterns between differentially expressed human papillomavirus (HPV) miRNAs and their significantly anti-correlated target protein-coding genes (PCGs) (p < 0.05) across all comparisons: (A) anal squamous cell carcinoma (ASCC), high-grade squamous intraepithelial lesions (HGSILs), and low-grade squamous intraepithelial lesions (LGSILs); (B) ASCC vs. HGSILs; (C) ASCC vs. LGSILs; and (D) ASCC vs. HGSILs + LGSILs. Empty circles denote HPV-miRNAs, while blue circles represent target genes. Edges indicate significant negative correlations, calculated using the Spearman correlation coefficient, with edge color intensity corresponding to the strength of the correlation. The ten most highly anti-correlated genes and the five HPV-derived miRNAs with the highest degree of connectivity are labeled in each network. For enhanced visualization and interactive exploration, these networks are shown in HTML versions of the figures, available at http://bioinfo.ipb.csic.es/Papers/ncRNA_HPV/, accessed on 1 June 2025.
Figure 4. Networks representing the correlation patterns between differentially expressed human papillomavirus (HPV) miRNAs and their significantly anti-correlated target protein-coding genes (PCGs) (p < 0.05) across all comparisons: (A) anal squamous cell carcinoma (ASCC), high-grade squamous intraepithelial lesions (HGSILs), and low-grade squamous intraepithelial lesions (LGSILs); (B) ASCC vs. HGSILs; (C) ASCC vs. LGSILs; and (D) ASCC vs. HGSILs + LGSILs. Empty circles denote HPV-miRNAs, while blue circles represent target genes. Edges indicate significant negative correlations, calculated using the Spearman correlation coefficient, with edge color intensity corresponding to the strength of the correlation. The ten most highly anti-correlated genes and the five HPV-derived miRNAs with the highest degree of connectivity are labeled in each network. For enhanced visualization and interactive exploration, these networks are shown in HTML versions of the figures, available at http://bioinfo.ipb.csic.es/Papers/ncRNA_HPV/, accessed on 1 June 2025.
Ncrna 11 00043 g004
Figure 5. Functional enrichment analysis of differentially expressed PCGs predicted to be target genes regulated by differentially expressed HPV-miRNAs. Ten most significantly enriched GO biological processes pathways ordered by gene ratios in the comparisons (A) anal squamous cell carcinoma (ASCC) vs. high-grade squamous intraepithelial lesions (HGSILs), (B) ASCC vs. low-grade squamous intraepithelial lesions (LGSILs), and (C) ASCC vs. HGSILs + LGSILs are shown. The size of the dots represents the number of predicted target genes in the gene list associated with the GO term and the color of the dots represents the p-value.
Figure 5. Functional enrichment analysis of differentially expressed PCGs predicted to be target genes regulated by differentially expressed HPV-miRNAs. Ten most significantly enriched GO biological processes pathways ordered by gene ratios in the comparisons (A) anal squamous cell carcinoma (ASCC) vs. high-grade squamous intraepithelial lesions (HGSILs), (B) ASCC vs. low-grade squamous intraepithelial lesions (LGSILs), and (C) ASCC vs. HGSILs + LGSILs are shown. The size of the dots represents the number of predicted target genes in the gene list associated with the GO term and the color of the dots represents the p-value.
Ncrna 11 00043 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

García, D.J.; Pulpillo-Berrocal, M.A.; Ruiz, J.L.; Andrés-León, E.; Terrón-Camero, L.C. Human Papillomavirus-Encoded microRNAs as Regulators of Human Gene Expression in Anal Squamous Cell Carcinoma: A Meta-Transcriptomics Study. Non-Coding RNA 2025, 11, 43. https://doi.org/10.3390/ncrna11030043

AMA Style

García DJ, Pulpillo-Berrocal MA, Ruiz JL, Andrés-León E, Terrón-Camero LC. Human Papillomavirus-Encoded microRNAs as Regulators of Human Gene Expression in Anal Squamous Cell Carcinoma: A Meta-Transcriptomics Study. Non-Coding RNA. 2025; 11(3):43. https://doi.org/10.3390/ncrna11030043

Chicago/Turabian Style

García, Daniel J., Marco A. Pulpillo-Berrocal, José L. Ruiz, Eduardo Andrés-León, and Laura C. Terrón-Camero. 2025. "Human Papillomavirus-Encoded microRNAs as Regulators of Human Gene Expression in Anal Squamous Cell Carcinoma: A Meta-Transcriptomics Study" Non-Coding RNA 11, no. 3: 43. https://doi.org/10.3390/ncrna11030043

APA Style

García, D. J., Pulpillo-Berrocal, M. A., Ruiz, J. L., Andrés-León, E., & Terrón-Camero, L. C. (2025). Human Papillomavirus-Encoded microRNAs as Regulators of Human Gene Expression in Anal Squamous Cell Carcinoma: A Meta-Transcriptomics Study. Non-Coding RNA, 11(3), 43. https://doi.org/10.3390/ncrna11030043

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop