Fact-checked by Grok 2 weeks ago

Piwi-interacting RNA

Piwi-interacting RNAs (piRNAs) are a class of small non-coding RNAs, typically 24–31 in length, that form ribonucleoprotein complexes with proteins—a subfamily of proteins—to primarily silence transposable elements and maintain integrity in animal germ cells. These RNAs are Dicer-independent, distinguishing them from microRNAs and small interfering RNAs, and are predominantly expressed in gonads such as testes and ovaries, though somatic expression occurs in certain species like arthropods and planarians. In mammals, piRNAs play crucial roles in by repressing selfish genetic elements that could otherwise disrupt and lead to sterility. The discovery of piRNAs dates back to 2006, when independent studies in and mouse germ cells identified these small RNAs as PIWI-associated molecules distinct from other regulatory RNAs, with key contributions from researchers including Alexei Aravin, Gregory Hannon, and Antoine Girard. Earlier work in the 1990s and early 2000s had identified genes in flies and mice as essential for development, but the associated RNAs were only characterized in the mid-2000s through deep sequencing of small RNA libraries from gonads. This breakthrough revealed piRNAs' abundance—millions of distinct sequences in some species—and their bias toward transposon-derived sequences, highlighting their role in an adaptive genome defense system. Biogenesis of piRNAs involves two main pathways: a primary pathway, where long precursor transcripts from genomic clusters are processed into mature piRNAs by endonucleases like (ZUC), and a secondary "ping-pong" amplification cycle that generates additional piRNAs through reciprocal slicing by -loaded complexes. In mammals, proteins such as PIWIL1 (MIWI), PIWIL2 (), PIWIL3, and PIWIL4 (MIWI2) orchestrate this process, with and MIWI2 mediating transcriptional silencing via of transposon loci during . Cofactors like GTSF1 and Tudor-domain proteins facilitate complex assembly and amplification, ensuring precise targeting of mobile elements. Beyond transposon repression, piRNAs contribute to broader gene regulation, mRNA stability, and even antiviral defense in species like mosquitoes, while recent studies have uncovered roles in somatic maintenance and tissue regeneration in planarians. In humans, dysregulation of the piRNA pathway is implicated in and certain cancers, underscoring its evolutionary conservation and therapeutic potential. Ongoing research, including ovary-specific pathways involving PIWIL3, continues to expand understanding of piRNA diversity across metazoans.

Characteristics and Discovery

Structural Features

Piwi-interacting RNAs (piRNAs) are a class of small non-coding RNAs typically 24–31 in length in , with species-specific variations such as 23–29 in Drosophila and 24–31 in mice. Unlike microRNAs or siRNAs, piRNAs are processed from single-stranded RNA precursors in a Dicer-independent manner, distinguishing them structurally and biogenetically from other small RNAs. A hallmark structural feature of mature piRNAs is a strong toward as the first at the 5' end (1U ), particularly evident in initiator piRNAs that guide transposon silencing; this arises from the sequence preferences of proteins, which favor targets beginning with . This 5' enrichment restricts the sequence space of piRNAs and facilitates their loading into PIWI-clade proteins. At the 3' end, piRNAs undergo 2'-O-, a catalyzed by the methyltransferase Hen1 (or its orthologs), which protects the RNA from degradation by exonucleases and enhances stability within the piRNA-induced silencing complex. This is essential for piRNA function, as unmethylated forms are rapidly degraded .

Historical Development

The PIWI subfamily of Argonaute proteins was first identified in 1997 through genetic screens in Drosophila melanogaster, where the Piwi gene was shown to be essential for maintaining germline stem cells in both males and females. Subsequent studies in 2002 revealed that PIWI homologs in Tetrahymena thermophila bind small RNAs to direct DNA elimination during genome rearrangement, suggesting a conserved role in RNA-guided nucleic acid modification. In 2003, profiling in identified a class of 24-29 RNAs derived primarily from repetitive sequences, termed repeat-associated small interfering RNAs (rasiRNAs), which accumulated in gonads and were implicated in silencing transposable elements. These rasiRNAs were later recognized as piRNAs, but at the time, their biogenesis and precise function remained unclear. Parallel work in other organisms hinted at similar pathways, but lacked direct association with proteins. The formal discovery of piRNAs occurred in 2006 through independent cloning efforts that identified 26-31 nucleotide RNAs specifically bound to proteins in mammalian germ cells. In , Aravin et al. described piRNAs associated with the protein (now known as PIWIL2) in testes, enriched for transposon sequences and accumulating during . Concurrently, Girard et al. reported a germline-specific class of small RNAs bound to MIWI (PIWIL1) and in and testes, while et al., Lau et al., and Watanabe et al. confirmed these findings across species, noting their abundance (over 50,000 unique sequences) and lack of dependence on for processing. In , Vagin et al. simultaneously established a distinct small RNA pathway involving , Aubergine, and Argonaute-3 proteins that silences transposons in the germline, linking rasiRNAs directly to function. These studies collectively coined the term "piRNA" for PIWI-interacting RNAs, distinguishing them from miRNAs and siRNAs by their length, 1U bias, and genomic origins. Subsequent milestones in 2007 elucidated piRNA amplification mechanisms, including the ping-pong cycle of mutually dependent primary and secondary piRNA production, as detailed by Brennecke et al. in Drosophila gonads, where piRNAs from discrete genomic clusters guide transposon silencing. This adaptive system was shown to respond to transposon activity, establishing piRNAs as key guardians of genome integrity in the germline. By the late 2000s, piRNA pathways were confirmed across metazoans, with high-impact studies expanding their roles beyond transposons to epigenetic regulation and development.

Genomic Organization

piRNA Loci

PiRNA loci are discrete genomic regions that serve as the primary sources of piRNA precursors, typically transcribed as long, non-coding RNAs that are subsequently processed into mature s. These loci are enriched in (TE) sequences and their degraded fragments, functioning as repositories of genetic information about past transposon invasions to enable targeted silencing in the and . In many , piRNA loci are organized into clusters, which can span tens to hundreds of kilobases and produce piRNAs predominantly from one strand (unistrand clusters) or both strands (dual-strand clusters). Unistrand clusters often feature dedicated promoters, splicing, and , allowing export via the Nxf1-Nxt1 machinery, while dual-strand clusters are more repetitive and heterochromatic. In , loci are exemplified by prominent clusters such as the dual-strand 42AB locus on chromosome 2R, which spans approximately 240 kb and generates germline s targeting mobile elements, and the unistrand (flam) locus on the , covering ~180 kb and primarily producing somatic s antisense to LTR retrotransposons like Gypsy family elements. These clusters are marked by and rely on the Rhino-Deadlock-Cutoff (RDC) complex for transcription initiation and maintenance. formation of such clusters can occur from repetitive transgenic sequences over generations, initiated by maternally inherited siRNAs that trigger biogenesis and modifications, though siRNAs become dispensable once the locus is established. Unistrand clusters like flam are evolutionarily conserved across the genus, emerging around 13-15 million years ago to suppress endogenous retroviruses (ERVs), with capturing env-containing TEs in antisense orientation. In nematodes like , loci (known as 21U-RNA loci) are predominantly clustered in two ~3 Mb repressive domains on chromosome IV, accounting for over 90% of production and enriched in marks that facilitate transcription. Each locus features an upstream Ruby motif (GTTTC) derived from snRNA promoters, which recruits , followed by downstream signals that induce polymerase pausing to generate ~28 nt precursors. In contrast, the related species Pristionchus pacificus exhibits a dispersed organization, with 88% of loci embedded in introns of active genes across autosomes (absent on the ) and associated with H3K36me3 active domains. This environment dictates expression mode, as demonstrated by perturbations that alter locus activity based on epigenetic context. Mammalian piRNA loci differ markedly, often lacking large dedicated and instead comprising thousands of dispersed sites, particularly for pachytene piRNAs produced during . Primary piRNAs derive from long single-stranded precursors transcribed from these loci, which are surveyed by the piRNA pathway to detect and silence transposons via and formation. In humans, piRNA loci show rapid divergence even among individuals, with syntenic conservation of cluster locations across mammals but sequence variability. In placental , piRNAs are predominantly expressed from single loci (51% of detected sequences) rather than clusters, with notable enrichment in the imprinted DLK1-DIO3 on 14q32.31, where 15 of 16 placenta-specific piRNAs map to the MEG8 . Additionally, in placental , 45% of known mitochondrial piRNA loci are placenta-expressed, highlighting tissue-specific .

Classification Schemes

An alternative classification scheme categorizes piRNAs by their genomic origins, highlighting functional diversity beyond transposon silencing. Transposon-derived piRNAs, the most abundant in cells, originate from (TE) sequences within piRNA clusters and directly target active TEs for silencing. Genic piRNAs derive from protein-coding gene 3' untranslated regions (3'UTRs), often in tissues, and may regulate mRNA stability or . Intergenic piRNAs, prevalent in mammalian testes, arise from non-genic regions and include pachytene piRNAs from precursors activated by transcription factors like A-MYB. Less common categories include those from long non-coding RNAs (lncRNAs) or bidirectional clusters, which contribute to broader epigenetic roles. This origin-based scheme underscores piRNA adaptability across species and cell types.

Biogenesis Pathways

Primary Pathway

The primary pathway of piRNA biogenesis generates the initial pool of piRNAs directly from genomic transcripts, primarily in germline cells, without requiring amplification from existing piRNAs. This pathway is conserved across species but exhibits variations in processing mechanisms and associated proteins. In Drosophila melanogaster, it begins with the transcription of long precursor RNAs from piRNA clusters, often bidirectional and enriched in transposon sequences, facilitated by chromatin regulators like Rhino, which binds H3K9me3 marks to promote cluster expression. Key processing steps involve nuclear export of precursors via factors such as UAP56, followed by cytoplasmic cleavage to define the 5' ends. The endonuclease (Zuc), a mitochondrial protein with activity, plays a central role in generating 5'-monophosphate termini on intermediates, independent of Dicer-like enzymes. These intermediates are then loaded onto proteins, where 3' ends are trimmed by exonucleases such as and Trimmer (PAPI-associated), and the mature (24-31 nt) undergo 2'-O-methylation at the 3' end by the methyltransferase Pimet (Hen1 in other species) for stability. In Drosophila gonads, this occurs in nuage-like structures or Yb bodies, with -loaded translocating to the for transposon silencing. Seminal work identified Zuc's endonucleolytic function through genetic screens and biochemical assays, establishing it as essential for primary production. In mammals, such as mice, the primary pathway similarly starts with transcription from piRNA clusters, but pachytene piRNAs predominate and derive from intergenic loci activated by the A-MYB during . Precursors are processed by the homolog MitoPLD (mitochondrial phospholipase D), which cleaves them into intermediates loaded onto proteins like (PIWIL2) or MIWI (PIWIL1). 3' ends are trimmed by the PNLDC1, followed by Henmt1-mediated . Unlike in flies, mammalian primary piRNAs show less toward transposons and extensive phasing, particularly for pachytene piRNAs, with processing occurring in nuage assemblies like chromatoid bodies. MitoPLD's role was confirmed through studies showing abolished piRNA production and defects. Across species, the primary pathway ensures a diverse repertoire for transposon surveillance, with loading onto specific clade proteins dictating downstream functions. Accessory proteins like (in flies) or MOV10L1 (in mammals) chaperone precursors, highlighting evolutionary adaptations while conserving core slicing and stabilization steps. Recent studies (as of 2025) have revealed that proteins facilitate biogenesis through inter-mitochondrial contacts and that Topoisomerase 3β aids in cytoplasmic precursor processing, enhancing efficiency across species.

Secondary Ping-Pong Cycle

The secondary ping-pong cycle represents a key mechanism in piRNA biogenesis, primarily operating in germ cells to boost the production of s that transposons for silencing. In this process, primary s, initially loaded into specific Piwi clade proteins, guide the endonucleolytic of complementary RNA transcripts from transposons or piRNA clusters. The 3' fragment from the is then captured and processed into a secondary , which is loaded into another protein and, in turn, directs the cleavage of additional transcripts, creating a reciprocal "ping-pong" loop that exponentially increases piRNA abundance against active transposons. In , the involves Aubergine (Aub) and 3 (Ago3) proteins localized in the nuage, a perinuclear structure in germ cells. Antisense primary piRNAs bound to Aub recognize and slice transposon mRNAs, generating a 5' fragment with a bias at position 1 (1U signature) that is trimmed and 2'-O-methylated before loading into Ago3. Ago3-bound piRNAs then cleave antisense transcripts from dual-strand piRNA clusters, producing new Aub-bound piRNAs with an adenosine bias at position 10 (10A signature), thus sustaining the amplification while linking post-transcriptional transposon silencing to further piRNA production. This heterotypic ping-pong requires accessory factors like Vasa and Krimper to assemble the complex and enforce strand bias, with endonuclease aiding 3' end formation of precursors. The is initiated by maternally deposited piRNAs or early primary piRNAs and is essential for adapting to transposon activity during . In mammals, such as mice, the ping-pong cycle operates similarly but involves MIWI-like (MILI) and MIWI2 proteins during embryonic prospermatogonia development. MILI-loaded antisense primary piRNAs cleave sense transposon transcripts (e.g., LINE1 elements), yielding secondary sense piRNAs with a 10A bias that load into MIWI2; these, in turn, slice antisense cluster transcripts to regenerate MILI-bound piRNAs with a 1U bias. This amplification directs MIWI2 to the , where it recruits factors for DNA methylation of transposon loci, establishing epigenetic silencing. Unlike in flies, mammalian ping-pong is phased, with cleavage products generating downstream piRNAs, and it relies on Tudor domain proteins like TDRD9 for complex assembly, though it diminishes in adult where non-ping-pong mechanisms predominate for pachytene piRNAs. The cycle's conservation across species underscores its role in transposon defense, though it is absent in nematodes like C. elegans, which use for amplification.

Phasing Mechanisms

Phasing mechanisms in piRNA biogenesis refer to the sequential processing of long precursor transcripts into multiple mature s, typically spaced at intervals of 24–32 , often initiated by a protein-mediated slicing event. This process amplifies piRNA production from a single precursor, ensuring efficient coverage of transposon sequences or other targets. Unlike the primary pathway, which generates discrete piRNAs from cluster transcripts, or the secondary ping-pong cycle, which amplifies sense-antisense pairs, phasing extends the output by cleaving the 3' fragment of a sliced precursor into a series of overlapping piRNAs with defined 5' ends. The is conserved across animals but shows species-specific variations in key effectors. In , phasing is prominently featured in the and cells, integrating with the ping-pong cycle. A PIWI-bound , such as those loaded onto Aubergine (Aub) or , guides slicing of a target transcript, generating a 5'-monophosphorylated 3' fragment. This fragment is then transported by the RNA helicase Armitage (Armi) to nuage structures or Yb bodies, where the endonuclease (Zuc) cleaves it at residues approximately every 26–28 to create the 5' ends of phased pre-piRNAs. The 3' ends are trimmed by exonucleases like Trimmer (PAPI/TDRD1) and , followed by 2'-O-methylation by Hen1 for stability, and loading onto for nuclear functions like transposon . Experimental evidence from Zuc mutants shows disrupted phasing, with reduced density and increased precursor accumulation, confirming Zuc's role in generating ~70% of Piwi-loaded piRNAs in gonads. This phased output often exhibits a 1U at the 5' end and overlaps of 10 nucleotides between consecutive piRNAs, reflecting the guide piRNA's slicing register. In mammals, particularly testes, phasing dominates pre-pachytene and pachytene piRNA production and relies on analogous but adapted components. proteins like MIWI2 (PIWIL4) or (PIWIL2) initiate slicing of cluster-derived transcripts, producing a 3' fragment that is processed at mitochondrial surfaces by the Zuc homolog MitoPLD/PLD6, which generates 5' ends in a phased manner. The MOV10L1 (Armi homolog) and Tudor proteins like TDRD5 facilitate precursor recruitment and ribosome-guided cleavage, enabling continuous 5'-to-3' progression along translated precursors up to 80 kb long. The 3' ends are refined by PNLDC1 trimming and HENMT1 before loading onto MIWI (PIWIL1). Unlike Drosophila, mammalian phasing is largely ping-pong independent for pachytene piRNAs and incorporates ribosomal scanning for precise spacing, as evidenced by deep sequencing showing phased piRNAs covering ~98% of cluster transcripts with minimal overlap. Mutations in PLD6 or MOV10L1 abolish phasing, leading to sterility and transposon derepression, underscoring its essentiality for . This mechanism has been unified across bilaterians by studies showing -directed slicing as the conserved trigger for all piRNA types.

Biological Functions

Transposon Silencing

Piwi-interacting RNAs (piRNAs) play a central role in transposon silencing, primarily in germlines, where they defend the against the mutagenic activity of transposable (TEs) by preventing their mobilization and integration. This silencing is essential for maintaining genomic stability during , as unchecked transposon activity can lead to sterility, developmental defects, or heritable mutations. piRNAs achieve this through interactions with clade proteins, which guide them to complementary transposon sequences, enabling both post-transcriptional and transcriptional repression mechanisms. In , piRNA-mediated transposon silencing operates via two complementary pathways. Post-transcriptional silencing occurs in the through the ping-pong amplification cycle, where Aubergine (Aub)-bound antisense piRNAs cleave sense transposon transcripts, generating sense piRNAs loaded into Argonaute 3 (Ago3); these in turn cleave antisense transcripts, amplifying the response with a characteristic 10-nucleotide overlap bias. This slicing activity directly degrades transposon mRNAs, as demonstrated in seminal studies identifying the piRNA pathway's role in suppressing hybrid dysgenesis caused by P-elements. Transcriptional silencing, mediated by nuclear Piwi-piRNA complexes, scans nascent transposon transcripts and recruits the PANDAS/SFiNX/PICTS complex, including factors like Rhino and , to deposit repressive marks via the methyltransferase SetDB1, leading to formation and locus repression. In mammals, such as mice, transposon silencing emphasizes epigenetic modifications during . Pre-pachytene piRNAs, produced from specialized clusters, load into PIWIL1 (MIWI) and PIWIL4 (MIWI2) to initiate a ping-pong cycle that amplifies TE-targeting piRNAs, primarily against and IAP retrotransposons. Nuclear MIWI2-piRNA complexes then direct DNA at transposon promoters via DNMT3A/3L, establishing heritable silencing that persists post-fertilization; this mechanism is critical for preventing transposon derepression in embryos. Unlike Drosophila, mammalian silencing relies less on RNA slicing and more on DNA , highlighting evolutionary adaptations in piRNA pathway components across species.

Antiviral Defense

Piwi-interacting RNAs (piRNAs) contribute to antiviral defense in select animal species, particularly by targeting viral genomes or transcripts through sequence-specific silencing mechanisms analogous to their role in transposon repression. This function extends beyond the in some cases, involving the production of virus-derived piRNAs (vpiRNAs) that guide proteins to cleave or repress RNA. While the piRNA pathway is not essential for antiviral immunity in all animals, evidence from and mammals highlights its adaptive role in restricting and integration. In vector mosquitoes such as , the pathway mounts a robust antiviral response against arboviruses like (DENV), (SINV), and chikungunya virus (CHIKV). Upon infection, viral is reverse-transcribed into episomal viral DNA (vDNA) by host retrotransposons, which is then transcribed into antisense strands to generate primary piRNAs. These load onto 4, a mosquito-specific Piwi protein, to initiate a ping-pong amplification cycle that produces vpiRNAs covering the viral , leading to 10- to 100-fold reduction in viral titers. Endogenous viral elements (EVEs) integrated into the mosquito further enhance this defense by producing constitutive antisense piRNAs that pre-emptively target incoming viruses, demonstrating an adaptive, heritable immunity. This mechanism operates independently of the primary (siRNA) pathway and is somatic, occurring in mosquito cells to limit transmission. In mammals, piRNAs provide defense against retroviral invasion, as exemplified by the (KoRV) in koalas (Phascolarctos cinereus). The piRNA response exhibits a biphasic pattern: an initial innate phase where unspliced proviral transcripts are processed into sense-biased piRNAs in the testis, degrading nascent viral RNA via Piwi-mediated slicing; followed by an adaptive phase where antisense piRNAs from genomic clusters silence integrated proviruses through epigenetic mechanisms. This pathway targets over 60 active KoRV insertions per individual, with piRNAs showing a strong ping-pong signature (10-nucleotide overlap bias) and covering the viral genome at high density. Similar EVE-derived piRNAs contribute to antiviral immunity in other mammals, such as mice, by repressing endogenous retroviruses (ERVs) that could reactivate as infectious agents. Notably, the piRNA pathway's antiviral role is context-specific; in adult , it is dispensable for defense against RNA viruses like Drosophila C virus, where the siRNA pathway predominates, though minor vpiRNA production occurs without functional impact. This variation underscores evolutionary adaptations of the piRNA system across species.

Epigenetic Regulation

Piwi-interacting RNAs (piRNAs) play a crucial role in epigenetic regulation, primarily by guiding PIWI proteins to specific genomic loci to induce heritable modifications that maintain stability and control , especially in cells. These modifications include and histone alterations, which silence transposable elements (TEs) and influence developmental processes. In mammals, piRNA-mediated epigenetic programming occurs during , where it establishes protective marks against TE invasion while preserving essential functions. A primary mechanism involves piRNAs directing de novo through proteins, such as MIWI2 in mice, which recruits DNA methyltransferases (DNMT3A/3L) to TE-rich regions during prospermatogonia development. This process methylates promoters of retrotransposons like and IAP, preventing their mobilization and ensuring integrity; disruptions in MIWI2 lead to derepression and sterility. Similarly, in , -piRNA complexes interact with heterochromatin protein 1a (HP1a) to promote H3K9 trimethylation (), fostering formation and transcriptional silencing of TEs at telomeric and pericentromeric loci. piRNAs also contribute to by establishing stable marks that persist across generations. In C. elegans, the ortholog PRG-1 binds piRNAs to initiate at foreign sequences, triggering secondary amplification and heritable silencing that can last multiple generations. In mice, pachytene piRNAs associated with MIWI regulate post-meiotic via 3' UTR targeting, indirectly influencing epigenetic states during the histone-to-protamine transition in spermatids. These mechanisms extend beyond TEs to ; for instance, piRNAs guide at the paternally imprinted Rasgrf1 locus, balancing allelic expression in offspring. In contexts, -PIWI pathways modulate epigenetic landscapes to regulate and . For example, in human cancers, aberrant expression recruits DNMTs to methylate tumor suppressor genes like PTEN, promoting oncogenesis through promoter hypermethylation. Histone-modifying complexes, such as those involving MLL3 for activation or SUV39H1 for repressive , are similarly guided by to alter accessibility at non-TE loci. These functions highlight piRNAs' versatility in epigenetic control, with implications for , , and .

Pathway Components

Piwi Proteins

Piwi proteins constitute a germline-enriched of the protein family, distinguished by their specific association with piwi-interacting RNAs (piRNAs) to form silencing complexes that defend animal genomes against transposon invasion. These proteins are essential for maintaining genomic integrity during , with mutations often leading to sterility and transposon derepression. Evolutionarily conserved across metazoans, proteins emerged as specialized effectors for transposon control, differing from other Argonautes that primarily bind microRNAs or siRNAs in contexts. Structurally, proteins adopt a conserved bilobed typical of Argonautes, comprising an N-terminal domain, a PAZ domain for anchoring the 3' end of guide RNAs, a MID domain for 5' end recognition, and a PIWI domain with RNase H-fold activity for target cleavage. In , the reveals flexible linkers (L0, L1, L2) connecting these domains, enabling conformational adaptability, along with a unique N-terminal extension stabilized by hydrophobic interactions. The MID domain coordinates a ion to bind the 5' phosphate, preferring a uridine bias at the first position, while the PAZ domain engages the 3' 2'-O-methylated end with micromolar affinity. Unlike canonical Argonautes with a , many proteins feature a modified in the PIWI domain, which in nuclear isoforms like abolishes slicer activity. Piwi proteins load piRNAs in a stepwise, chaperone-mediated involving and Tudor-domain proteins, forming stable complexes that guide target recognition via base-pairing. Cytoplasmic Piwi paralogs, such as mouse (PIWIL1) and (PIWIL2), retain endonuclease activity to cleave transposon transcripts, initiating piRNA amplification cycles, whereas nuclear forms like (PIWIL4) direct formation without slicing. This functional dichotomy is enhanced by accessories like GTSF1, which boosts slicing efficiency in cytoplasmic complexes. Across species, proteins exhibit paralog diversification: has three (Piwi, Aubergine, Ago3), with Piwi localized to nuclei for transcriptional silencing and the others in cytoplasmic nuage for post-transcriptional control. Mammals encode four PIWIL proteins, primarily in testes (PIWIL1–2,4) or ovaries (PIWIL3 in select species), reflecting adaptations to sex-specific demands. These variations underscore Piwi proteins' core role in piRNA-directed transposon repression while enabling species-specific regulations, such as translational activation in spermiogenesis.

Tudor Domain Proteins

Tudor domain proteins are a family of scaffolding factors that play essential roles in piRNA biogenesis by binding to symmetrically dimethylated arginine (sDMA) residues on proteins, thereby facilitating the assembly of multiprotein complexes in the . These proteins, characterized by one or more domains that recognize sDMA modifications catalyzed by the methyltransferase PRMT5, are conserved across and are crucial for transposon silencing during . In the piRNA pathway, they promote the efficiency of primary piRNA production and the ping-pong amplification cycle, often localizing to cytoplasmic structures such as nuage and Yb bodies. In , Tudor domain proteins are integral to both somatic and germline . Vreteno (Vret), a multi-Tudor protein, acts as a core scaffold in the primary piRNA pathway, interacting with , , and to support piRNA precursor processing in Yb bodies and nuage; its depletion leads to a collapse in piRNA populations and transposon derepression, resulting in sterility. The Tdrd12 homologs—such as Yb (soma-specific), Brother of Yb (BoYb), and Sister of Yb (SoYb)—enhance primary piRNA biogenesis by stabilizing PIWI complexes and facilitating Piwi nuclear import; Yb mutants exhibit reduced ping-pong signatures and transposon activation in follicle cells. In the ping-pong cycle, proteins like Krimper (Krimp) and Qin/Kumo selectively promote Aubergine (Aub)-Ago3 heterotypic interactions by binding sDMAs on Aub, ensuring phased piRNA amplification and transposon targeting; Krimp mutants show defective Ago3 loading and impaired secondary piRNA production. Additionally, (Tej) and Spn-E contribute to nuage assembly and piRNA intermediate handover, underscoring the coordinated action of proteins in maintaining pathway robustness. In mammals, Tudor domain proteins exhibit analogous scaffolding functions but with adaptations for species-specific piRNA dynamics. TDRD1, containing multiple extended Tudor domains, serves as a molecular scaffold for MIWI and MILI proteins, binding sDMAs with varying affinities (e.g., 35 μM for its TD3 domain) to assemble biogenesis complexes in nuage-like structures; Tdrd1 mice display spermatogenic arrest and transposon derepression due to disrupted piRNA loading. TDRD12 is specifically required for secondary piRNA production in mice, interacting with MILI via its second Tudor domain in a methylation-independent manner to load piRNAs onto MIWI2 for nuclear transposon silencing; Tdrd12 mutants show reduced MIWI2 nuclear localization, decreased of LINE1 and IAP elements (from ~90% to 53-61%), and . TDRD5 further enhances the ping-pong cycle by binding piRNA precursors and promoting MILI-Aub-like amplification, with its Tudor domains facilitating selective interactions in spermatocytes. Across species, these proteins ensure piRNA pathway fidelity by preventing off-target amplification and supporting epigenetic regulation, highlighting their evolutionary conservation in germline protection.

Other Accessory Proteins

In the piRNA pathway, accessory proteins play essential roles in precursor transcription, nuclear export, processing, loading onto Piwi proteins, and stabilization, distinct from the core Piwi and Tudor domain components. These proteins facilitate the specificity and efficiency of piRNA-mediated transposon silencing in germline cells across species. In Drosophila melanogaster, nuclear factors such as Rhino (Rhi), a heterochromatin protein 1 (HP1) homolog, bind to H3K9me3-marked dual-strand piRNA clusters to promote non-canonical transcription of precursors, enabling their incorporation into the pathway. Deadlock associates with Rhino to support this transcriptional activation, while Cutoff (Cuff), a transcription termination cofactor, protects the 5′ ends of precursors from degradation, ensuring sufficient substrate for processing. UAP56 (Hel25E), an RNA helicase, mediates nuclear export of these precursors by interacting with Rhino at cluster loci. Cytoplasmic processing relies on proteins like (Zuc), a mitochondrial endonuclease that cleaves single-stranded precursors to generate mature 5′ ends, a step critical for primary biogenesis. (Armi), a non-DEAD-box , facilitates loading of primary into proteins and is required for nuage localization, where processing occurs. Vasa (Vas), a DEAD-box RNA , supports the ping-pong amplification cycle by unwinding RNA duplexes and promoting Aubergine-mediated slicing. Maelstrom (Mael), featuring an XPD-like motif and an RNase H-like domain, is essential for silencing; it localizes to nuage, promotes ping-pong cycle integrity by binding sliced intermediates, and enforces transcriptional repression of transposons, with mutants showing derepression and sterility. Stabilization is achieved by Pimet (Hen1), an RNA methyltransferase that adds a 2′--methyl group to 3′ ends, enhancing resistance to exonucleases. In mammals, analogous accessory proteins adapt the pathway to pachytene and pre-pachytene piRNA production. MOV10L1, a DExD/H-box , is indispensable for precursor processing and associates with all Piwi proteins (MIWI, , MIWI2) to generate mature piRNAs, with its depletion causing meiotic arrest and transposon derepression. MitoPLD (PLD6), the homolog, exhibits endonuclease activity on mitochondrial surfaces to trim piRNA intermediates, essential for nuage assembly and fertility. GPAT2, a mitochondrial outer , interacts with to support primary piRNA biogenesis in prospermatogonia. The A-MYB (MYBL1) drives expression of pachytene piRNA precursors and pathway genes in round spermatids, linking hormonal cues to piRNA production. (MAEL), conserved from flies, localizes to nuage-like structures, binds piRNA-Piwi complexes, and is required for transposon repression and ; biallelic variants in MAEL cause human male infertility through piRNA biogenesis defects and transposon derepression, as reported in studies up to 2024. These accessory proteins often localize to specialized structures like nuage or mitochondria, integrating piRNA biogenesis with cellular architecture, and their conservation underscores the pathway's evolutionary robustness despite species-specific adaptations.

Species-Specific Variations

In Drosophila

Piwi-interacting RNAs (piRNAs) were first identified in the germline of Drosophila melanogaster, where they were characterized as 24–29 nucleotide small RNAs bound to PIWI clade Argonaute proteins, distinct from miRNAs and siRNAs due to their lack of dependence on Dicer enzymes. Their discovery stemmed from small RNA cloning efforts in the early 2000s, revealing abundant germline-specific RNAs associated with the Piwi protein, initially named repeat-associated small interfering RNAs (rasiRNAs) before being redesignated piRNAs in 2006. In Drosophila, piRNAs primarily function to silence transposable elements (TEs) in gonads, protecting genome integrity during gametogenesis, with mutations in piRNA pathway components leading to sterility from TE derepression and DNA damage. Biogenesis of piRNAs in Drosophila occurs through phased processing of long precursor transcripts from discrete genomic loci called piRNA clusters, which are enriched in TE sequences and can span up to 200 kb. In the germline, dual-strand clusters (e.g., 42AB) are transcribed by RNA polymerase II under control of the Rhino-Deadlock-Cutoff (RDC) complex, which promotes heterochromatin formation and read-through transcription despite repressive marks like H3K9me3. Primary piRNAs are generated in the cytoplasm via endonucleolytic cleavage by Zucchini (Zuc), a mitochondrial endoribonuclease, followed by 3' end trimming by Nibbler and 2'-O-methylation by Hen1 for stability; these load onto Aubergine (Aub) or Piwi. A hallmark of the Drosophila germline pathway is the ping-pong amplification cycle, where Aub-loaded antisense piRNAs cleave sense TE transcripts to produce sense piRNAs that load into Argonaute 3 (Ago3), which in turn cleaves cluster transcripts to regenerate antisense piRNAs, amplifying the response and exhibiting a characteristic 10-nucleotide overlap bias. This cycle localizes to nuage granules, electron-dense perinuclear structures involving Vasa helicase and Tudor-domain proteins. In gonadal cells, such as cells, piRNA production follows a distinct pathway using uni-strand clusters like (flam), a >180 kb locus on the densely packed with 104 insertions from 42 TE families, producing predominantly antisense piRNAs to target specific TEs like gypsy. The locus, discovered through genetic studies tracing back to linking it to gypsy regulation, exemplifies -specific TE surveillance, with piRNAs loading exclusively onto for nuclear import and transcriptional silencing via deposition by histone methyltransferases. Somatic biogenesis is Zuc-dependent but lacks ping-pong amplification, instead relying on phasing from Zuc cleavage sites in Yb-body organelles, involving factors like , Yb, and Gasz; this pathway silences TEs post-transcriptionally in nurse cells and transcriptionally in follicle cells. Unlike the , somatic piRNAs show strand bias and are tuned to somatic TE activity, preventing ectopic expression during . A unique feature of piRNA regulation in Drosophila is paramutation-like transgenerational inheritance, where maternally deposited piRNAs from the ping-pong cycle prime to silence paternally inherited s, enabling rapid adaptation to new TE invasions without immediate evolution. This is mediated by Aub- and Ago3-bound piRNAs in the , which direct nascent piRNA production and marking. Seminal work identified piRNA clusters in 2007, revealing their role as "piRNA factories," and the ping-pong mechanism in the same year, establishing Drosophila as the model for piRNA-mediated TE defense across animals.00043-4) Compared to mammals, Drosophila piRNAs exhibit greater reliance on amplification cycles and specialized nuclear complexes like RDC, absent in vertebrates, highlighting ary adaptations in fly germline protection.

In Mammals and Other Organisms

In mammals, piwi-interacting RNAs (piRNAs) are predominantly expressed in the gonads, where they play a critical role in safeguarding genomic integrity during gametogenesis, particularly by silencing transposable elements (TEs) in the germline. Unlike in Drosophila, mammalian piRNA biogenesis features distinct phases: pre-pachytene piRNAs, derived from TE-rich clusters, are amplified through a ping-pong cycle involving the PIWI proteins MILI (PIWIL2) and MIWI2 (PIWIL4), which generate 26–28 nucleotide piRNAs with a characteristic 10A bias; these guide post-transcriptional cleavage of TE transcripts by MILI and nuclear recruitment of de novo DNA methyltransferases (DNMT3A/DNMT3L) by MIWI2 for transcriptional silencing. Pachytene piRNAs, produced later in spermatogenesis from euchromatic, uni-strand clusters transcribed by RNA polymerase II under A-MYB regulation, are longer (∼30 nt) and primarily non-TE-derived, associating with MIWI (PIWIL1) to fine-tune gene expression and reinforce TE repression. This pathway is essential for male fertility; disruptions, such as in Spocd1 knockout mice, lead to derepression of young TEs like LINE1 and IAP, causing DNA damage, meiotic arrest, and infertility. A key mechanistic innovation in mammals is the "two-factor authentication" for precise targeting during embryonic prospermatogonia stages (E13.5–E14.5), where SPIN1 reads bivalent marks (/) on nascent promoters to recruit SPOCD1, priming before MIWI2-piRNA complexes engage transcripts and direct DNMT3A/L via TDRD5 and TEX15 scaffolding. In mice, this ensures of ∼70% of young TEs, preventing their mobilization during the epigenetic reprogramming of primordial germ cells. piRNA profiles mirror this, with testis-specific expression of PIWIL1–4 and dynamic clusters producing both pre- and post-pachytene piRNAs, though piRNA detection remains controversial and likely artifactual in non-gonadal tissues. In female mammals, piRNAs are less abundant but support oocyte maturation; for instance, MIWI2 guides TE in growing s, with deficiencies linked to ovarian defects. Beyond mammals, piRNA pathways exhibit species-specific adaptations in other organisms, reflecting diverse evolutionary pressures on TE control. In zebrafish (Danio rerio), piRNAs operate in both germline and soma, with Zwiwi (PIWIL1 ortholog) binding 26–29 nt piRNAs to silence TEs via slicer activity, extending protection to embryonic development and stress responses, unlike the gonad-restricted expression in mammals. Caenorhabditis elegans lacks a ping-pong cycle, instead generating 21U-RNAs (piRNA analogs) from ∼15,000 monoallelic genomic loci via a TOFU/PEL complex, which trigger nuclear Argonaute (CSR-1/PRG-1) pathways for TE silencing and gene licensing through 22G-RNA amplification by RNA-dependent RNA polymerases (RdRPs). In planarians (Schmidtea mediterranea), piRNAs from uni-strand clusters (with minimal ping-pong signatures) support stem cell pluripotency and regeneration, targeting TEs in neoblasts to maintain genome stability during tissue remodeling. These variations highlight how piRNA systems adapt to organismal biology, prioritizing DNA methylation in mammals, slicer-mediated cleavage in fish, and RdRP amplification in nematodes.

Research Advances

Experimental Methods

Experimental methods for studying piwi-interacting RNAs (piRNAs) primarily involve high-throughput sequencing, biochemical assays, and genetic manipulations to identify, profile, and characterize their biogenesis and functions. These techniques have evolved to distinguish piRNAs from other small RNAs, leveraging their unique length (24-31 nucleotides), 2'-O-methylation at the 3' end, and association with proteins. Early identification relied on (IP) of proteins followed by sequencing of bound small RNAs, which confirmed piRNA enrichment in tissues across species like and mice. Small RNA sequencing (sRNA-seq) remains the cornerstone for piRNA detection and profiling, enabling genome-wide analysis of piRNA expression in tissues such as testes, ovaries, and somatic cells. Libraries are prepared from total , size-selected for 24-31 nt fragments, and sequenced using next-generation platforms; bioinformatics pipelines then filter reads by mapping to piRNA clusters and excluding miRNA/siRNA annotations. This approach has profiled over 1,400 piRNA datasets from 114 metazoan species, revealing tissue-specific and transposon-targeting piRNAs. Recent 2025 computational advances, including deep learning-based piRNA target prediction models, have improved accuracy in identifying piRNA-mRNA interactions across species. Complementary techniques like quantitative reverse transcription (qRT-PCR) and northern blotting validate specific piRNA levels, while 3' end methylation-specific assays confirm piRNA identity. To study piRNA-protein interactions and targets, crosslinking immunoprecipitation sequencing (CLIP-seq) and immunoprecipitation sequencing (RIP-seq) capture PIWI-piRNA complexes or associated transcripts. In CLIP-seq, UV crosslinking stabilizes interactions, followed by RNase digestion, with anti-PIWI antibodies, and sequencing of protected fragments, which maps piRNA binding sites on transposons or mRNAs. RIP-seq simplifies this by omitting crosslinking, focusing on co-precipitated RNAs, and has identified piRNA targets in cancer cells. Crosslinking, ligation, and sequencing of hybrids (CLASH) further reveals direct piRNA-target duplexes, as demonstrated in for validating slicing activity. Biochemical assays elucidate piRNA biogenesis and effector functions, particularly the ping-pong amplification cycle central to secondary piRNA production. In vitro slicer assays reconstitute PIWI-mediated cleavage of target RNAs, using recombinant proteins and radiolabeled substrates to measure endonuclease activity, often enhanced by domain co-factors. The (Zuc) endonuclease assay, involving IP of Zuc from extracts and monitoring cleavage of piRNA precursors, dissects primary piRNA processing in nuage bodies. (LNA) inhibitors or antisense oligonucleotides target specific piRNAs to assess functional impacts, such as transposon derepression or changes via reporter assays. Genetic approaches in model organisms provide insights into pathways. In , CRISPR/Cas9-mediated knockouts of genes (e.g., Aubergine, ) or piRNA cluster deletions reveal defects in transposon silencing and fertility, quantified by dysgenic sterility rates and genomic instability metrics. (RNAi) screens in cell lines like Schneider 2 cells identify pathway components by monitoring piRNA levels post-knockdown. In mice, conditional knockouts of MIWI proteins using Cre-loxP systems demonstrate epigenetic roles in , with piRNA reductions measured by sRNA-seq. These methods, often combined with for / or xenograft models for disease contexts, establish piRNA's conserved roles while highlighting species-specific variations.

Emerging Roles and Implications

Recent research has revealed that piRNAs extend beyond their canonical role in transposon silencing within cells to influence functions and disease pathogenesis. In tissues, piRNA/ complexes contribute to antiviral defense, as seen in mosquitoes where they target non-retroviral RNA viruses, thereby restricting viral replication. In neoblasts, these complexes regulate and maintain integrity during tissue regeneration and differentiation, highlighting their involvement in biology and . Additionally, piRNAs participate in suppression, which has implications for and evolutionary dynamics by preventing biased transmission of genetic elements. In cancer, aberrant piRNA expression promotes tumorigenesis through transcriptional and post-transcriptional mechanisms, including and mRNA decay. For instance, in gastric cancer, piR-651 promotes by inhibiting G2/M phase arrest, while piR-823 acts as a tumor suppressor by inhibiting . In , piR-1245 correlates with poor prognosis by enhancing , and piR-54265 boosts proliferation via PIWI interactions. Similar patterns emerge in gynecological cancers; piR-33733 and piR-52207 are overexpressed in , regulating migration and invasion through pathways like TGF-β and Wnt/β-catenin. In , piR-55490 represses to curb growth, whereas piR-651 promotes it, underscoring piRNAs' dual oncogenic and suppressive roles. These dysregulations often involve epigenetic modifications such as , positioning piRNAs as key regulators of cancer progression. piRNAs also play emerging roles in non-oncological diseases, particularly in cardiovascular and respiratory conditions. In , CFRPi (piRNA-000691) degrades mRNA, exacerbating via the PI3K-AKT-mTOR pathway, while piRNA-6426 alleviates by targeting SOAT1 through DNMT3B-mediated silencing. In respiratory diseases like , piR-020381 and piR-020490 serve as biomarkers for latent by modulating pathways, and in , piRNAs such as DQ596390 influence PTEN signaling in cells. For pulmonary arterial , piR-63076 promotes via Acadm promoter . The therapeutic and diagnostic implications of piRNAs are profound, with their expression profiles offering potential as biomarkers for early detection and . In gastrointestinal cancers, panels including piR-823 and piR-10506469 predict outcomes and could guide personalized therapies. Targeting proteins, such as HIWI knockdown to inhibit growth or antagomirs against CFRPi for cardiac , represents promising strategies to mitigate progression. In gynecological malignancies, modulating piR-17560 could overcome chemoresistance, emphasizing piRNAs' translational potential. As of 2025, piRNAs have shown promise as therapeutics in via targeted delivery and in modulating for . Ongoing studies continue to elucidate these mechanisms, paving the way for piRNA-based interventions in diverse pathologies.

References

  1. [1]
    PIWI-interacting RNAs: who, what, when, where, why, and how - NIH
    Sep 26, 2024 · PIWI-interacting RNAs (piRNA) suppress selfish genetic elements and are essential for germ cell biology in animals.
  2. [2]
  3. [3]
  4. [4]
  5. [5]
  6. [6]
  7. [7]
  8. [8]
    Unistrand piRNA clusters are an evolutionarily conserved ... - Nature
    Nov 13, 2023 · The PIWI-interacting RNA (piRNA) pathway prevents endogenous genomic parasites, i.e. transposable elements, from damaging the genetic ...
  9. [9]
  10. [10]
  11. [11]
    how mammalian piRNAs are produced, originated, and evolved - PMC
    PIWI-interacting RNAs (piRNAs) are small non-coding RNAs that form 1:1 RNA–protein complexes with PIWI (P-element-induced wimpy testis) proteins. The PIWI gene ...Summary Of Pirna Biogenic... · Table 2 · Location For Pirna...
  12. [12]
    Human placental piwi-interacting RNA transcriptome is ... - Nature
    Jul 22, 2021 · Placenta-derived piRNA are primarily derived from single loci. As the organization of genomic regions where piRNA are located can be suggestive ...
  13. [13]
  14. [14]
  15. [15]
    Biology of PIWI-interacting RNAs: new insights into biogenesis and ...
    These small RNAs can be classified on the basis of their origins, processing factors, and Argonaute-binding partners (Kim et al. 2009).Missing: schemes | Show results with:schemes
  16. [16]
    piRNAs: from biogenesis to function - Company of Biologists journals
    Sep 15, 2014 · Several studies simultaneously reported the identification of Piwi-interacting RNAs from mouse and rat germ cells (Aravin et al., 2006; Girard ...
  17. [17]
    Mammalian piRNAs: Biogenesis, function, and mysteries - PMC
    During primary processing, the genomic loci that encode piRNAs are initially transcribed as long RNAs, termed piRNA precursors, that can be tens to hundreds of ...Introduction · Pirna Biogenesis · Piwi-Associated Proteins
  18. [18]
  19. [19]
  20. [20]
  21. [21]
    A Single Mechanism of Biogenesis, Initiated and Directed by PIWI ...
    A single model that explains piRNA production in most animals. piRNAs initiate piRNA production by guiding PIWI proteins to slice precursor transcripts.
  22. [22]
    piRNA processing within non‐membrane structures is governed by ...
    Dec 30, 2024 · Here, we focuses on the well-studied biogenesis of pre-pachytene piRNAs, which involves phasing processing and ping-pong amplification. For ...
  23. [23]
    An introduction to PIWI-interacting RNAs (piRNAs) in the context of ...
    Oct 10, 2022 · PiRNAs are small RNAs associated with PIWI proteins, part of an adaptive immune system that restricts mobile genetic elements, originating from ...
  24. [24]
    piRNA-Guided Transposon Silencing and Response to Stress ... - NIH
    Apr 30, 2024 · Here, we review transposon regulation by piRNAs and the piRNA pathway regulation in response to stress, focusing on the Drosophila female germline.
  25. [25]
  26. [26]
  27. [27]
  28. [28]
  29. [29]
  30. [30]
  31. [31]
  32. [32]
  33. [33]
  34. [34]
  35. [35]
    Crystal structure of Drosophila Piwi | Nature Communications
    Feb 12, 2020 · Here, we report the crystal structure of the Drosophila PIWI-clade Argonaute Piwi in complex with endogenous piRNAs, at 2.9 Å resolution.
  36. [36]
    PIWI-interacting RNA (piRNA): a narrative review of its biogenesis ...
    The present narrative review aims to introduce the details of piRNA biogenesis and function and then detail the various discoveries as a timeline of events. ...
  37. [37]
    How does the Royal Family of Tudor rule the PIWI-interacting RNA ...
    This mechanism helps to maintain the integrity of the genome and the development of gametes. PIWI proteins have been shown recently to contain symmetrical ...
  38. [38]
    A systematic analysis of Drosophila TUDOR domain-containing ...
    Recent studies linked the piRNA pathway to TUDOR biology as TUDOR domains of various proteins bind symmetrically methylated Arginine residues in PIWI proteins.
  39. [39]
    One Loop to Rule Them All: The Ping-Pong Cycle and piRNA ...
    Discovered nearly a decade ago, this small RNA silencing system comprises PIWI-clade Argonaute proteins and their associated RNA-binding partners, the piRNAs.Missing: history papers
  40. [40]
    The multiple Tudor domain-containing protein TDRD1 is a molecular ...
    Tudor domain-containing 1 (TDRD1) is an interaction partner of mouse Piwi proteins and is implicated in piRNA biogenesis.Missing: seminal papers
  41. [41]
    Tudor domain containing 12 (TDRD12) is essential for ... - PNAS
    We find that TDRD12 is dispensable for primary piRNA biogenesis but essential for production of secondary piRNAs that enter Piwi protein MIWI2 (PIWIL4).
  42. [42]
    TDRD5 binds piRNA precursors and selectively enhances ... - Nature
    Jan 9, 2018 · Tudor domain proteins play conserved roles in regulating the piRNA pathway and spermatogenesis by interacting with the PIWI proteins. Tudor ...
  43. [43]
  44. [44]
  45. [45]
  46. [46]
  47. [47]
  48. [48]
    piRNA Biogenesis in Drosophila Melanogaster - PubMed Central
    In this review, we elaborate on piRNA biogenesis in Drosophila somatic and germline cells. We focus on the mechanisms by which piRNA precursor transcription is ...Missing: original | Show results with:original
  49. [49]
    piRNAs—the ancient hunters of genome invaders
    In their initial efforts to characterize miRNAs, Aravin et al. (2003) cloned total small RNA populations from different developmental stages of Drosophila. In ...<|separator|>
  50. [50]
    The piRNA pathway in Drosophila ovarian germ and somatic cells
    PiRNAs are small non-coding RNAs that repress transposons, forming piRISCs to protect the germline genome. They are germline-specific and crucial for genome ...Figure 1 · Figure 2 · Figure 3
  51. [51]
    History of the discovery of a master locus producing piRNAs - NIH
    Aug 4, 2014 · The discovery of transposable elements (TEs) in the 1950s by B. McClintock implied the existence of cellular regulatory systems controlling ...Missing: seminal papers
  52. [52]
    The emergence of piRNAs against transposon invasion to preserve ...
    Nov 10, 2017 · This review features our current understandings of mammalian PIWI-interacting RNAs (piRNAs) and their role in TE regulation in spermatogenesis.Primary Pirna Biogenesis · Pre-Pachytene Pirnas... · Pachytene Pirnas Regulate...
  53. [53]
  54. [54]
    How mammalian piRNAs instruct de novo DNA methylation ... - Nature
    Sep 7, 2020 · This research provided the first mechanistic insights into how piRNA directs de novo DNA methylation in male mammals.Missing: review | Show results with:review
  55. [55]
    Two-factor authentication underpins the precision of the piRNA ...
    Sep 18, 2024 · The PIWI-interacting RNA (piRNA) pathway guides the DNA methylation of young, active transposons during germline development in male mice.
  56. [56]
  57. [57]
  58. [58]
  59. [59]
  60. [60]
    The biogenesis and biological function of PIWI-interacting RNA in ...
    Jun 12, 2021 · PIWI-interacting RNAs (piRNAs) are a novel type of small ncRNA initially discovered in germ cells that have a specific length (24–31 nucleotides) ...Missing: definition | Show results with:definition
  61. [61]
  62. [62]
    PIWI-interacting RNAs: who, what, when, where, why, and how | The EMBO Journal
    ### Summary of Experimental Methods for Studying piRNAs (from https://www.embopress.org/doi/10.1038/s44318-024-00253-8)
  63. [63]
    The Emerging Role of PIWI-Interacting RNAs (piRNAs) in ... - MDPI
    This review describes important topics about piRNAs including their molecular characteristics, biosynthesis processes, gene expression silencing mechanisms, and ...
  64. [64]
    A comprehensive review on the role of PIWI-interacting RNA (piRNA ...
    Nov 15, 2024 · piRNAs are classified into three groups based on their origin: transposon-derived piRNAs, mRNA-derived piRNAs, and lncRNA-derived piRNAs [6].
  65. [65]
    The emerging role of the piRNA/PIWI complex in respiratory tract ...
    Mar 13, 2023 · In this review, we discuss current studies summarizing the biogenetic processes, functions, and emerging roles of piRNAs in respiratory tract diseases.
  66. [66]