Piwi-interacting RNA

Last updated

Piwi-interacting RNA (piRNA) is the largest class of small non-coding RNA molecules expressed in animal cells. [1] [2] [3] piRNAs form RNA-protein complexes through interactions with piwi-subfamily Argonaute proteins. These piRNA complexes are mostly involved in the epigenetic and post-transcriptional silencing of transposable elements and other spurious or repeat-derived transcripts, but can also be involved in the regulation of other genetic elements in germ line cells. [4] [5] [6]

Contents

piRNAs are mostly created from loci that function as transposon traps which provide a kind of RNA-mediated adaptive immunity against transposon expansions and invasions. [7] They are distinct from microRNA (miRNA) in size (26–31 nucleotides as opposed to 21–24 nt), lack of sequence conservation, increased complexity, and independence of Dicer for biogenesis, at least in animals. [5] [1] [2] (Plant Dcl2 may play a role in rasi/piRNA biogenesis.) [8] [9]

Double-stranded RNAs capable of silencing repeat elements, then known as repeat associated small interfering RNA (rasiRNA), were proposed in Drosophila in 2001. [10] By 2008, it was still unclear how piRNAs are generated, but potential methods had been suggested, and it was certain their biogenesis pathway is distinct from miRNA and siRNA, while rasiRNA is now considered a piRNA subspecies. [11]

Characteristics

Proposed piRNA structure, with the 3' end 2'-O-methylation PiRNA.jpg
Proposed piRNA structure, with the 3′ end 2′-O-methylation

piRNAs have been identified in both vertebrates and invertebrates, and although biogenesis and modes of action do vary somewhat between species, a number of features are conserved. piRNAs have no clear secondary structure motifs, [1] [12] due to the fact that the length of a piRNA varies between species (from 21 to 31 nucleotides), and the bias for a 5’ uridine is common to piRNAs in both vertebrates and invertebrates. piRNAs in Caenorhabditis elegans have a 5’ monophosphate and a 3’ modification that acts to block either the 2’ or 3’ oxygen; [13] this has also been confirmed to exist in Drosophila melanogaster , [14] zebrafish, [15] mice, [16] and rats. [15] This 3’ modification is a 2’-O-methylation; the reason for this modification is not clear, but it has been suggested that it increases piRNA stability. [15] [17]

More than 50,000 unique piRNA sequences have been discovered in mice and more than 13,000 in D. melanogaster. [18] It is thought that there are many hundreds of thousands of different piRNA species in mammals. [19]

History and loci

In the early 1980s, it was discovered that a single mutation in the fruit fly genome could specifically activate all copies of a retrovirus-like element called Gypsy in the female germline. The site of the mutations that made these Gypsies "dance" was thus called the flamenco locus. In 2001, Aravin et al. proposed that double-stranded (ds) RNA-mediated silencing is implicated in the control of retrotransposons in the germline and by 2003 the idea had emerged that vestiges of transposons might produce dsRNAs required for the silencing of "live" transposons. [10] Sequencing of the 200,000-bp flamenco locus was difficult, as it turned out to be packed with transposable element fragments (104 insertions of 42 different transposons, including multiple Gypsies), all facing the same direction. Indeed, piRNAs are all found in clusters throughout animal genomes; these clusters may contain as few as ten or many thousands of piRNAs matching different, phased transposon fragments. This led to the idea in 2007 that in germlines a pool of primary piRNAs is processed from long single-stranded transcripts encoded by piRNA clusters in the opposite orientation of the transposons, so that the piRNAs can anneal to and complement the transposon-encoded transcripts, thereby triggering their degradation. Any transposon landing in the correct orientation in such a cluster will make the individual more or less immune to that transposon, and such an advantageous mutation will spread quickly through the population. The original mutations in the flamenco locus inhibited the transcription of the master transcript, thereby deactivating this defense system. [7] [20] [1] [21] [22]

A historical example of invasion and Piwi response is known: the P-element transposon invaded a Drosophila melanogaster genome in the mid-20th century, and, through interbreeding, within decades all wild fruit flies worldwide (though not the reproductively isolated lab strains) contained the same P-element. Repression of further P-element activity, spreading near-simultaneously, appears to have occurred by the Piwi-interacting RNA pathway. [23]

piRNA clusters in genomes can now readily be detected via bioinformatics methods. [24] While D. melanogaster and vertebrate piRNAs have been located in areas lacking any protein-coding genes, [11] [20] piRNAs in C. elegans have been identified amidst protein-coding genes. [13]

In mammals, piRNAs are found both in testes [25] and ovaries, [26] although they only seem to be required in males. [4] In invertebrates, piRNAs have been detected in both the male and female germlines. [15] [19]

At the cellular level, piRNAs have been found within both the nucleus and cytoplasm, suggesting that piRNA pathways may function in both of these areas [11] and, therefore, may have multiple effects. [27]

Classification

There are at least three Argonaute (Ago) subfamilies that have been found in eukaryotes. Unlike the Ago subfamily which is present in animals, plants, and fission yeast, the Piwi subfamily has only been found in animals. [28] RasiRNA has been observed in Drosophila and some unicellular eukaryotes but its presence in mammals has not been determined, unlike piRNA which has been observed in many species of invertebrates and vertebrates including mammals; [29] however, since proteins which associate with rasiRNA are found in both vertebrates and invertebrates, it is possible that active rasiRNA exist and have yet to be observed in other animals. RasiRNAs have been observed in Schizosaccharomyces pombe , a species of yeast, as well in some plants, neither of which have been observed to contain the Piwi subfamily of Argonaute proteins. [8] It has been observed that both rasiRNA and piRNA are maternally linked, but more specifically it is the Piwi protein subfamily that is maternally linked and therefore leads to the observation that rasiRNA and piRNA are maternally linked.[ clarification needed ] [30]

Biogenesis

The ping-pong mechanism for the biogenesis of the 5' end of rasiRNA. RasiRNA biogen.jpg
The ping-pong mechanism for the biogenesis of the 5′ end of rasiRNA.

The biogenesis of piRNAs is not yet fully understood, although possible mechanisms have been proposed. piRNAs show a significant strand bias, that is, they are derived from one strand of DNA only, [1] and this may indicate that they are the product of long single stranded precursor molecules. [2] A primary processing pathway is suggested to be the only pathway used to produce pachytene piRNAs; in this mechanism, piRNA precursors are transcribed resulting in piRNAs with a tendency to target 5’ uridines. [31] [32] Also proposed is a ‘Ping Pong’ mechanism wherein primary piRNAs recognise their complementary targets and cause the recruitment of piwi proteins. This results in the cleavage of the transcript at a point ten nucleotides from the 5’ end of the primary piRNA, producing the secondary piRNA. [32] These secondary piRNAs are targeted toward sequences that possess an adenine at the tenth position. [31] Since the piRNA involved in the ping pong cycle directs its attacks on transposon transcripts, the ping pong cycle acts only at the level of transcription. [22] One or both of these mechanisms may be acting in different species; C. elegans , for instance, does have piRNAs, but does not appear to use the ping pong mechanism at all. [19]

A significant number of piRNAs identified in zebrafish and D. melanogaster contain adenine at their tenth position, [11] and this has been interpreted as possible evidence of a conserved biosynthetic mechanism across species. [17] Ping-pong signatures have been identified in very primitive animals such as sponges and cnidarians, pointing to the existence of the ping-pong cycle already in the early branches of metazoans. [33]

Ping Pong

The piRNA Ping-Pong pathway was first proposed from studies in Drosophila where the piRNA associated with the two cytoplasmic Piwi proteins, Aubergine (Aub) and Argonaute-3 (Ago3) exhibited a high frequency of sequence complementarity over exactly 10 nucleotides at their 5′ ends. [32] [34] This relationship is known as the "ping-pong signature" and is also observed in associated piRNA from Mili and Miwi2 proteins isolated from mouse testes. The proposed function of Ping-Pong in Drosophila or in mouse remains to be understood, but a leading hypothesis is that the interaction between Aub and Ago3 allows for a cyclic refinement of piRNA that are best suited to target active transposon sequences. Aub piRNA are primarily antisense to transposable element transcripts and are believed to be the main factor in targeting deleterious transcripts through complementarity. Conversely, Ago3 piRNA sequences are predominantly of sense orientation to transposable element transcripts and are derived from the product of Aub cleavage of transposon mRNA. As such, Ago3 piRNA lack the ability to target transposable element transcripts directly. Therefore, it was proposed that Ago3 piRNA guide the production of piRNA that are loaded into Aub by targeting newly exported piRNA cluster transcripts. Several lines of evidence support the effect of Ago3 on the production of Aub piRNA, in particular from examining the piRNA repertoire in Drosophila ovaries that are mutant for Ago3 and the Tudor-domain protein Kumo/Qin. [35] [36]

The molecular mechanism that underpins Ping-Pong likely involves several piRNA pathway associated factors. Qin was reported to coordinate the loading of Ago3 with piRNA, in addition to interacting with both Aub and Ago3. [36] However, the Tudor protein krimper ( A1ZAC4 ) was also shown to interact with both Aub and Ago3 through its Tudor domains while also binding itself through its N-terminal Krimper domain. [37] Specifically, Krimper interacts with Ago3 in its piRNA-unloaded state, while its interaction with Aub is dependent on the symmetrical dimethylation of arginine residues in the N-terminal region of Aub. [37] [38] In Silkmoth germ cells, it was proposed that Vasa protein coordinates the Ping-Pong mechanism of Silkmoth Aub (Siwi) and Ago3. [39]

It is likely that the mechanism of Ping-Pong is primarily coordinated by Krimper but factors such as Kumo/Qin and Vasa, in addition to other factors have necessary functions in the Ping-Pong mechanism.

piRNA Phasing

The Drosophila piRNA pathway can be separated into two branches: the cytoplasmic branch consisting of Aub and Ago3 operating the Ping-Pong mechanism, and the nuclear branch, pertaining to the co-transcriptional silencing of genomic loci by Piwi in the nucleus. Through complementary strategies, two studies show that Aub and Ago3 target cleavage triggers the 'phased' loading of piRNA into Piwi. [40] [41] Phasing begins with the targeting and cleavage of a complementary target by either Aub or Ago3 associated with a 'responder' piRNA. Once cleaved, the targeted transcript is then processed further by a mechanism believed to require the mitochondrial-associated endonuclease, Zucchini, which leads to the loading of Piwi protein with sequential fragments of the targeted transcript. In this way, the Aub or Ago3 'responder' piRNA sequence cleaves a complementary target that is then sliced at periodic intervals of approximately 27 nucleotides that are sequentially loaded into Piwi protein. Once loaded with piRNA, Piwi then enters the germ cell nucleus to co-transcriptionally silence nascent transcripts with complementarity to its piRNA guide. [42] It is currently unknown whether phasing occurs in other organisms.

Function

The wide variation in piRNA sequences and piwi function across species contributes to the difficulty in establishing the functionality of piRNAs. [43] However, like other small RNAs, piRNAs are thought to be involved in gene silencing, [1] specifically the silencing of transposons. [44] The majority of piRNAs are antisense to transposon sequences, [3] [22] suggesting that transposons are targets of the piRNAs. In mammals, it appears that the activity of piRNAs in transposon silencing is most important during the development of the embryo, [31] and in both C. elegans and humans, piRNAs are necessary for spermatogenesis. [43]

RNA silencing

piRNA has a role in RNA silencing via the formation of an RNA-induced silencing complex (RISC). piRNAs interact with piwi proteins that are part of a family of proteins called the Argonautes. These are active in the testes of mammals and are required for germ-cell and stem-cell development in invertebrates. Three piwi subfamily proteins – MIWI, MIWI2, and MILI – have been found to be essential for spermatogenesis in mice. piRNAs direct the piwi proteins to their transposon targets. [31] A decrease or absence of PIWI gene expression is correlated with an increased expression of transposons. [11] [31] Transposons have a high potential to cause deleterious effects on their hosts [21] and, in fact, mutations in piRNA pathways have been found to reduce fertility in D. melanogaster. [20] Further, it is thought that piRNA and endogenous small interfering RNA (endo-siRNA) may have comparable and even redundant functionality in transposon control in mammalian oocytes. [22]

piRNAs appear to affect particular methyltransferases that perform the methylations which are required to recognise and silence transposons, [31] but this relationship is not well understood.

Antiviral effects

In Dipterans viral-derived piRNAs derived from positive-sense RNA viruses were first identified in Drosophila ovarian somatic sheet (OSS) cells. [45] Subsequent experimental studies have demonstrated that the piRNA pathway is not required for antiviral defence in Drosophila melanogaster. [46] However, in mosquitoes the PIWI family of proteins has expanded [47] and some PIWI proteins have been identified as antiviral such as Piwi4. [48] As such virus infections in mosquitoes commonly produce virus-derived piRNAs in diverse positive-sense RNA, [49] negative-sense RNA [50] [48] and single-stranded DNA viruses. [51]

Epigenetic effects

piRNAs can be transmitted maternally, [15] and based on research in D. melanogaster, piRNAs may be involved in maternally derived epigenetic effects. [20] The activity of specific piRNAs in the epigenetic process also requires interactions between piwi proteins and HP1a, as well as other factors. [18]

Accessory proteins of the piRNA pathway

Genetic screens examining fertility defects identified a number of proteins that are not Piwi-clade Argonautes, yet produce the same sterility phenotypes as Piwi mutants.

Drosophila Tudor domain proteins

Many factors required for the piRNA pathway in Drosophila contain Tudor domains that are known to bind symmetrically dimethylated arginine residues (sDMA) present in methylation motifs of Piwi proteins. Piwi proteins are symmetrically dimethylated by the PRMT5 methylosome complex, consisting of Valois (MEP50) and Capsulèen (dart5; PRMT5). [52] [53]

Non-Tudor Drosophila piRNA pathway proteins

Drosophila nuclear piRNA pathway proteins

Investigation

Major advances in the study of piRNA have been achieved thanks to the use of next-generation sequencing techniques, such as Solexa, 454, and Illumina platform sequencing. These techniques allow analysis of highly complex and heterogeneous RNA populations like piRNAs. Due to their small size, expression and amplification of small RNAs can be challenging, so specialised PCR-based methods have been developed in response to this difficulty. [54] [55] However, research has also revealed that a number of annotated piRNAs may be false positives; for instance, a majority of piRNAs that were expressed in somatic non-gonadal tissues were considered to derive from non-coding RNA fragments. [56]

Related Research Articles

<span class="mw-page-title-main">Transposable element</span> Semiparasitic DNA sequence

A transposable element is a nucleic acid sequence in DNA that can change its position within a genome, sometimes creating or reversing mutations and altering the cell's genetic identity and genome size. Transposition often results in duplication of the same genetic material. In the human genome, L1 and Alu elements are two examples. Barbara McClintock's discovery of them earned her a Nobel Prize in 1983. Its importance in personalized medicine is becoming increasingly relevant, as well as gaining more attention in data analytics given the difficulty of analysis in very high dimensional spaces.

Small RNA (sRNA) are polymeric RNA molecules that are less than 200 nucleotides in length, and are usually non-coding. RNA silencing is often a function of these molecules, with the most common and well-studied example being RNA interference (RNAi), in which endogenously expressed microRNA (miRNA) or exogenously derived small interfering RNA (siRNA) induces the degradation of complementary messenger RNA. Other classes of small RNA have been identified, including piwi-interacting RNA (piRNA) and its subspecies repeat associated small interfering RNA (rasiRNA). Small RNA "is unable to induce RNAi alone, and to accomplish the task it must form the core of the RNA–protein complex termed the RNA-induced silencing complex (RISC), specifically with Argonaute protein".

<span class="mw-page-title-main">Dicer</span> Enzyme that cleaves double-stranded RNA (dsRNA) into short dsRNA fragments

Dicer, also known as endoribonuclease Dicer or helicase with RNase motif, is an enzyme that in humans is encoded by the DICER1 gene. Being part of the RNase III family, Dicer cleaves double-stranded RNA (dsRNA) and pre-microRNA (pre-miRNA) into short double-stranded RNA fragments called small interfering RNA and microRNA, respectively. These fragments are approximately 20–25 base pairs long with a two-base overhang on the 3′-end. Dicer facilitates the activation of the RNA-induced silencing complex (RISC), which is essential for RNA interference. RISC has a catalytic component Argonaute, which is an endonuclease capable of degrading messenger RNA (mRNA).

<span class="mw-page-title-main">Retrotransposon</span> Type of genetic component

Retrotransposons are a type of genetic component that copy and paste themselves into different genomic locations (transposon) by converting RNA back into DNA through the reverse transcription process using an RNA transposition intermediate.

The RNA-induced silencing complex, or RISC, is a multiprotein complex, specifically a ribonucleoprotein, which functions in gene silencing via a variety of pathways at the transcriptional and translational levels. Using single-stranded RNA (ssRNA) fragments, such as microRNA (miRNA), or double-stranded small interfering RNA (siRNA), the complex functions as a key tool in gene regulation. The single strand of RNA acts as a template for RISC to recognize complementary messenger RNA (mRNA) transcript. Once found, one of the proteins in RISC, Argonaute, activates and cleaves the mRNA. This process is called RNA interference (RNAi) and it is found in many eukaryotes; it is a key process in defense against viral infections, as it is triggered by the presence of double-stranded RNA (dsRNA).

<span class="mw-page-title-main">Hop (protein)</span> Protein-coding gene in the species Homo sapiens

Hop, occasionally written HOP, is an abbreviation for Hsp70-Hsp90 Organizing Protein. It functions as a co-chaperone which reversibly links together the protein chaperones Hsp70 and Hsp90.

<span class="mw-page-title-main">Argonaute</span> Protein that plays a role in RNA silencing process

The Argonaute protein family, first discovered for its evolutionarily conserved stem cell function, plays a central role in RNA silencing processes as essential components of the RNA-induced silencing complex (RISC). RISC is responsible for the gene silencing phenomenon known as RNA interference (RNAi). Argonaute proteins bind different classes of small non-coding RNAs, including microRNAs (miRNAs), small interfering RNAs (siRNAs) and Piwi-interacting RNAs (piRNAs). Small RNAs guide Argonaute proteins to their specific targets through sequence complementarity, which then leads to mRNA cleavage, translation inhibition, and/or the initiation of mRNA decay.

<span class="mw-page-title-main">Piwi</span> Genes and regulatory proteins

Piwi genes were identified as regulatory proteins responsible for stem cell and germ cell differentiation. Piwi is an abbreviation of P-elementInduced WImpy testis in Drosophila. Piwi proteins are highly conserved RNA-binding proteins and are present in both plants and animals. Piwi proteins belong to the Argonaute/Piwi family and have been classified as nuclear proteins. Studies on Drosophila have also indicated that Piwi proteins have no slicer activity conferred by the presence of the Piwi domain. In addition, Piwi associates with heterochromatin protein 1, an epigenetic modifier, and piRNA-complementary sequences. These are indications of the role Piwi plays in epigenetic regulation. Piwi proteins are also thought to control the biogenesis of piRNA as many Piwi-like proteins contain slicer activity which would allow Piwi proteins to process precursor piRNA into mature piRNA.

RNA silencing or RNA interference refers to a family of gene silencing effects by which gene expression is negatively regulated by non-coding RNAs such as microRNAs. RNA silencing may also be defined as sequence-specific regulation of gene expression triggered by double-stranded RNA (dsRNA). RNA silencing mechanisms are conserved among most eukaryotes. The most common and well-studied example is RNA interference (RNAi), in which endogenously expressed microRNA (miRNA) or exogenously derived small interfering RNA (siRNA) induces the degradation of complementary messenger RNA. Other classes of small RNA have been identified, including piwi-interacting RNA (piRNA) and its subspecies repeat associated small interfering RNA (rasiRNA).

<span class="mw-page-title-main">Tudor domain</span>

In molecular biology, a Tudor domain is a conserved protein structural domain originally identified in the Tudor protein encoded in Drosophila. The Tudor gene was found in a Drosophila screen for maternal factors that regulate embryonic development or fertility. Mutations here are lethal for offspring, inspiring the name Tudor, as a reference to the Tudor King Henry VIII and the several miscarriages experienced by his wives.

Nuage are Drosophila melanogaster germline granules. Nuage are the hallmark of Drosophila melanogaster germline cells, which have an electron-dense perinuclear structure and can silence the selfish genetic elements in Drosophila melanogaster. The term 'Nuage' comes from the French word for 'cloud', as they appear as nebulous electron-dense bodies by electron microscopy. They are found in nurse cells of the developing Drosophila melanogaster egg chamber and are composed of various types of proteins, including RNA-helicases, Tudor domain proteins, Piwi-clade Argonaute proteins, in addition to a PRMT5 methylosome composed of Capsuléen and its co-factor, Valois (MEP50).

Transposon silencing is a form of transcriptional gene silencing targeting transposons. Transcriptional gene silencing is a product of histone modifications that prevent the transcription of a particular area of DNA. Transcriptional silencing of transposons is crucial to the maintenance of a genome. The “jumping” of transposons generates genomic instability and can cause extremely deleterious mutations. Transposable element insertions have been linked to many diseases including hemophilia, severe combined immunodeficiency, and predisposition to cancer. The silencing of transposons is therefore extremely critical in the germline in order to stop transposon mutations from developing and being passed on to the next generation. Additionally, these epigenetic defenses against transposons can be heritable. Studies in Drosophila, Arabidopsis thaliana, and mice all indicate that small interfering RNAs are responsible for transposon silencing. In animals, these siRNAS and piRNAs are most active in the gonads.

RDE-1 (RNAi-DEfective 1) is a primary Argonaute protein required for RNA-mediated interference (RNAi) in Caenorhabditis elegans. The rde-1 gene locus was first characterized in C. elegans mutants resistant to RNAi, and is a member of a highly conserved Piwi gene family that includes plant, Drosophila, and vertebrate homologs.

The gene Maelstrom, Mael, creates a protein, which was first located in Drosophila melanogaster in the nuage perinuclear structure and has functionality analogous to the spindle, spn, gene class. Its mammalian homolog is MAEL.

<span class="mw-page-title-main">Retrotransposon silencing</span>

Transposable elements are pieces of genetic material that are capable of splicing themselves into a host genome and then self propagating throughout the genome, much like a virus. Retrotransposons are a subset of transposable elements that use an RNA intermediate and reverse transcribe themselves into the genome. Retrotransposon proliferation may lead to insertional mutagenesis, disrupt the process of DNA repair, or cause errors during chromosomal crossover, and so it is advantageous for an organism to possess the means to suppress or "silence" retrotransposon activity.

Ruth Lehmann is a developmental and cell biologist. She is the Director of the Whitehead Institute for Biomedical Research. She previously was affiliated with the New York University School of Medicine, where she was the Director of the Skirball Institute of Biomolecular Medicine, the Laura and Isaac Perlmutter Professor of Cell Biology, and the Chair of the Department of Cell Biology. Her research focuses on germ cells and embryogenesis.

piwi-interacting RNA (piRNA) belongs to the small RNA class found in eukaryotic organisms, their major role is to regulate the expression of genes by the mRNA degradation or silencing. After the association with argonaute protein family, these short RNA guide the RNA-induced silencing complex (RISC) to their target by sequence complementarity. piRNAs have approximately from 26 to 31 nucleotides and are found in almost all metazoans.

Haifan Lin is a Chinese-born American stem cell biologist. He is the Eugene Higgins Chair Professor of Cell Biology at Yale University and the founding Director of the Yale Stem Cell Center. He previously founded and directed the Stem Cell Research Program at Duke University. Recognized for his significant contributions to stem cell research, he was elected to the US National Academy of Sciences and American Academy of Arts and Sciences in 2018.

<span class="mw-page-title-main">Neoblast</span> Planarian regeneration proliferative cells

Neoblasts (ˈniːəʊˌblæst) are adult stem cells found in planarian flatworms. They are the only dividing planarian cells, and they produce all cell types, including the germline. Neoblasts are abundant in the planarian parenchyma, and comprise up to 30 percent of all cells. Following injury, neoblasts rapidly divide and generate new cells, which allows planarians to regenerate any missing tissue.

References

  1. 1 2 3 4 5 6 "Molecular Biology Select". Cell. 126 (2): 223–225. July 2006. doi: 10.1016/j.cell.2006.07.012 .
  2. 1 2 3 Seto AG, Kingston RE, Lau NC (June 2007). "The coming of age for Piwi proteins". Molecular Cell. 26 (5): 603–609. doi: 10.1016/j.molcel.2007.05.021 . PMID   17560367.
  3. 1 2 Monga I, Banerjee I (November 2019). "Computational Identification of piRNAs Using Features Based on RNA Sequence, Structure, Thermodynamic and Physicochemical Properties". Current Genomics. 20 (7): 508–518. doi:10.2174/1389202920666191129112705. PMC   7327968 . PMID   32655289.
  4. 1 2 Siomi MC, Sato K, Pezic D, Aravin AA (April 2011). "PIWI-interacting small RNAs: the vanguard of genome defence". Nature Reviews Molecular Cell Biology. 12 (4): 246–258. doi:10.1038/nrm3089. PMID   21427766. S2CID   5710813.
  5. 1 2 Dorner S, Eulalio A, Huntzinger E, Izaurralde E (August 2007). "Delving into the diversity of silencing pathways. Symposium on MicroRNAs and siRNAs: biological functions and mechanisms". EMBO Reports. 8 (8): 723–729. doi:10.1038/sj.embor.7401015. PMC   1978081 . PMID   17599087.
  6. Klattenhoff C, Bratu DP, McGinnis-Schultz N, Koppetsch BS, Cook HA, Theurkauf WE (January 2007). "Drosophila rasiRNA pathway mutations disrupt embryonic axis specification through activation of an ATR/Chk2 DNA damage response". Developmental Cell. 12 (1): 45–55. doi: 10.1016/j.devcel.2006.12.001 . PMID   17199040.
  7. 1 2 Goriaux C, Théron E, Brasset E, Vaury C (2014). "History of the discovery of a master locus producing piRNAs: the flamenco/COM locus in Drosophila melanogaster". Frontiers in Genetics. 5: 257. doi: 10.3389/fgene.2014.00257 . PMC   4120762 . PMID   25136352.
  8. 1 2 Aravin A, Tuschl T (October 2005). "Identification and characterization of small RNAs involved in RNA silencing". FEBS Letters. 579 (26): 5830–5840. doi: 10.1016/j.febslet.2005.08.009 . PMID   16153643.
  9. Xie Z, Johansen LK, Gustafson AM, Kasschau KD, Lellis AD, Zilberman D, Jacobsen SE, Carrington JC (May 2004). "Genetic and functional diversification of small RNA pathways in plants". PLOS Biology. 2 (5): E104. doi: 10.1371/journal.pbio.0020104 . PMC   350667 . PMID   15024409.
  10. 1 2 Aravin AA, Naumova NM, Tulin AV, Vagin VV, Rozovsky YM, Gvozdev VA (July 2001). "Double-stranded RNA-mediated silencing of genomic tandem repeats and transposable elements in the D. melanogaster germline". Current Biology. 11 (13): 1017–1027. Bibcode:2001CBio...11.1017A. doi: 10.1016/S0960-9822(01)00299-8 . PMID   11470406. S2CID   14767819.
  11. 1 2 3 4 5 Klattenhoff C, Theurkauf W (January 2008). "Biogenesis and germline functions of piRNAs". Development. 135 (1): 3–9. doi: 10.1242/dev.006486 . PMID   18032451.
  12. Carmen L, Michela B, Rosaria V, Gabriella M (2009). "Existence of snoRNA, microRNA, piRNA characteristics in a novel non-coding RNA: x-ncRNA and its biological implication in Homo sapiens". Journal of Bioinformatics and Sequence Analysis. 1 (2): 031–040.
  13. 1 2 Ruby JG, Jan C, Player C, Axtell MJ, Lee W, Nusbaum C, Ge H, Bartel DP (December 2006). "Large-scale sequencing reveals 21U-RNAs and additional microRNAs and endogenous siRNAs in C. elegans". Cell. 127 (6): 1193–1207. doi: 10.1016/j.cell.2006.10.040 . PMID   17174894. S2CID   16838469.
  14. Vagin VV, Sigova A, Li C, Seitz H, Gvozdev V, Zamore PD (July 2006). "A distinct small RNA pathway silences selfish genetic elements in the germline". Science. 313 (5785): 320–324. Bibcode:2006Sci...313..320V. doi:10.1126/science.1129333. PMID   16809489. S2CID   40471466.
  15. 1 2 3 4 5 Houwing S, Kamminga LM, Berezikov E, Cronembold D, Girard A, van den Elst H, et al. (April 2007). "A role for Piwi and piRNAs in germ cell maintenance and transposon silencing in Zebrafish". Cell. 129 (1): 69–82. doi:10.1016/j.cell.2007.03.026. hdl: 11858/00-001M-0000-0012-E169-6 . PMID   17418787. S2CID   13373509.
  16. Kirino Y, Mourelatos Z (April 2007). "Mouse Piwi-interacting RNAs are 2′-O-methylated at their 3′ termini". Nature Structural & Molecular Biology. 14 (4): 347–348. doi:10.1038/nsmb1218. PMID   17384647. S2CID   31193964.
  17. 1 2 Faehnle CR, Joshua-Tor L (October 2007). "Argonautes confront new small RNAs". Current Opinion in Chemical Biology. 11 (5): 569–577. doi:10.1016/j.cbpa.2007.08.032. PMC   2077831 . PMID   17928262.
  18. 1 2 Lin H, Yin H, Beyret E, Findley S, Deng W (2008). "The role of the piRNA pathway in stem cell self-renewal". Developmental Biology. 319 (2): 479. doi:10.1016/j.ydbio.2008.05.048.
  19. 1 2 3 Das PP, Bagijn MP, Goldstein LD, Woolford JR, Lehrbach NJ, Sapetschnig A, Buhecha HR, Gilchrist MJ, Howe KL, Stark R, Matthews N, Berezikov E, Ketting RF, Tavaré S, Miska EA (July 2008). "Piwi and piRNAs act upstream of an endogenous siRNA pathway to suppress Tc3 transposon mobility in the Caenorhabditis elegans germline". Molecular Cell. 31 (1): 79–90. doi:10.1016/j.molcel.2008.06.003. PMC   3353317 . PMID   18571451.
  20. 1 2 3 4 Brennecke J, Malone CD, Aravin AA, Sachidanandam R, Stark A, Hannon GJ (November 2008). "An epigenetic role for maternally inherited piRNAs in transposon silencing". Science. 322 (5906): 1387–1392. Bibcode:2008Sci...322.1387B. doi:10.1126/science.1165171. PMC   2805124 . PMID   19039138.
  21. 1 2 O'Donnell KA, Boeke JD (April 2007). "Mighty Piwis defend the germline against genome intruders". Cell. 129 (1): 37–44. doi:10.1016/j.cell.2007.03.028. PMC   4122227 . PMID   17418784.
  22. 1 2 3 4 Malone CD, Hannon GJ (February 2009). "Small RNAs as guardians of the genome". Cell. 136 (4): 656–668. doi:10.1016/j.cell.2009.01.045. PMC   2792755 . PMID   19239887.
  23. Kelleher ES (August 2016). "Reexamining the P-Element Invasion of Drosophila melanogaster Through the Lens of piRNA Silencing". Genetics. 203 (4): 1513–1531. doi:10.1534/genetics.115.184119. PMC   4981261 . PMID   27516614.
  24. Rosenkranz D, Zischler H (January 2012). "proTRAC—a software for probabilistic piRNA cluster detection, visualization and analysis". BMC Bioinformatics. 13 (5): 5. doi: 10.1186/1471-2105-13-5 . PMC   3293768 . PMID   22233380.
  25. Aravin A, Gaidatzis D, Pfeffer S, Lagos-Quintana M, Landgraf P, Iovino N, Morris P, Brownstein MJ, Kuramochi-Miyagawa S, Nakano T, Chien M, Russo JJ, Ju J, Sheridan R, Sander C, Zavolan M, Tuschl T (July 2006). "A novel class of small RNAs bind to MILI protein in mouse testes". Nature. 442 (7099): 203–207. Bibcode:2006Natur.442..203A. doi:10.1038/nature04916. PMID   16751777. S2CID   4379895.
  26. Tam OH, Aravin AA, Stein P, Girard A, Murchison EP, Cheloufi S, Hodges E, Anger M, Sachidanandam R, Schultz RM, Hannon GJ (May 2008). "Pseudogene-derived small interfering RNAs regulate gene expression in mouse oocytes". Nature. 453 (7194): 534–538. Bibcode:2008Natur.453..534T. doi:10.1038/nature06904. PMC   2981145 . PMID   18404147.
  27. Ruvkun G (July 2008). "Tiny RNA: Where do we come from? What are we? Where are we going?". Trends in Plant Science. 13 (7): 313–316. doi: 10.1016/j.tplants.2008.05.005 . PMID   18562240.
  28. Houwing S, Kamminga LM, Berezikov E, Cronembold D, Girard A, van den Elst H, Filippov DV, Blaser H, Raz E, Moens CB, Plasterk RH, Hannon GJ, Draper BW, Ketting RF (April 2007). "A role for Piwi and piRNAs in germ cell maintenance and transposon silencing in Zebrafish". Cell. 129 (1): 69–82. doi:10.1016/j.cell.2007.03.026. hdl: 11858/00-001M-0000-0012-E169-6 . PMID   17418787. S2CID   13373509.
  29. Girard A, Sachidanandam R, Hannon GJ, Carmell MA (July 2006). "A germline-specific class of small RNAs binds mammalian Piwi proteins". Nature. 442 (7099): 199–202. Bibcode:2006Natur.442..199G. doi:10.1038/nature04917. PMID   16751776. S2CID   3185036.
  30. Tomari Y, Du T, Haley B, Schwarz DS, Bennett R, Cook HA, Koppetsch BS, Theurkauf WE, Zamore PD (March 2004). "RISC assembly defects in the Drosophila RNAi mutant armitage". Cell. 116 (6): 831–841. doi: 10.1016/S0092-8674(04)00218-1 . PMID   15035985. S2CID   17588448.
  31. 1 2 3 4 5 6 Aravin AA, Sachidanandam R, Bourc'his D, Schaefer C, Pezic D, Toth KF, Bestor T, Hannon GJ (September 2008). "A piRNA pathway primed by individual transposons is linked to de novo DNA methylation in mice". Molecular Cell. 31 (6): 785–799. doi:10.1016/j.molcel.2008.09.003. PMC   2730041 . PMID   18922463.
  32. 1 2 3 Brennecke J, Aravin AA, Stark A, Dus M, Kellis M, Sachidanandam R, Hannon GJ (March 2007). "Discrete small RNA-generating loci as master regulators of transposon activity in Drosophila" (PDF). Cell. 128 (6): 1089–1103. doi: 10.1016/j.cell.2007.01.043 . PMID   17346786. S2CID   2246942.
  33. Grimson A, Srivastava M, Fahey B, Woodcroft BJ, Chiang HR, King N, Degnan BM, Rokhsar DS, Bartel DP (October 2008). "Early origins and evolution of microRNAs and Piwi-interacting RNAs in animals". Nature. 455 (7217): 1193–1197. Bibcode:2008Natur.455.1193G. doi:10.1038/nature07415. PMC   3837422 . PMID   18830242.
  34. Gunawardane LS, Saito K, Nishida KM, Miyoshi K, Kawamura Y, Nagami T, Siomi H, Siomi MC (March 2007). "A slicer-mediated mechanism for repeat-associated siRNA 5′ end formation in Drosophila". Science. 315 (5818): 1587–1590. Bibcode:2007Sci...315.1587G. doi: 10.1126/science.1140494 . PMID   17322028. S2CID   11513777.
  35. Li C, Vagin VV, Lee S, Xu J, Ma S, Xi H, Seitz H, Horwich MD, Syrzycka M, Honda BM, Kittler EL, Zapp ML, Klattenhoff C, Schulz N, Theurkauf WE, Weng Z, Zamore PD (May 2009). "Collapse of germline piRNAs in the absence of Argonaute3 reveals somatic piRNAs in flies". Cell. 137 (3): 509–521. doi:10.1016/j.cell.2009.04.027. PMC   2768572 . PMID   19395009.
  36. 1 2 Zhang Z, Xu J, Koppetsch BS, Wang J, Tipping C, Ma S, Weng Z, Theurkauf WE, Zamore PD (November 2011). "Heterotypic piRNA Ping-Pong requires qin, a protein with both E3 ligase and Tudor domains". Molecular Cell. 44 (4): 572–584. doi:10.1016/j.molcel.2011.10.011. PMC   3236501 . PMID   22099305.
  37. 1 2 Webster A, Li S, Hur JK, Wachsmuth M, Bois JS, Perkins EM, Patel DJ, Aravin AA (August 2015). "Aub and Ago3 Are Recruited to Nuage through Two Mechanisms to Form a Ping-Pong Complex Assembled by Krimper". Molecular Cell. 59 (4): 564–575. doi:10.1016/j.molcel.2015.07.017. PMC   4545750 . PMID   26295961.
  38. Sato K, Iwasaki YW, Shibuya A, Carninci P, Tsuchizawa Y, Ishizu H, Siomi MC, Siomi H (August 2015). "Krimper Enforces an Antisense Bias on piRNA Pools by Binding AGO3 in the Drosophila Germline". Molecular Cell. 59 (4): 553–563. doi: 10.1016/j.molcel.2015.06.024 . PMID   26212455.
  39. Xiol J, Spinelli P, Laussmann MA, Homolka D, Yang Z, Cora E, Couté Y, Conn S, Kadlec J, Sachidanandam R, Kaksonen M, Cusack S, Ephrussi A, Pillai RS (June 2014). "RNA clamping by Vasa assembles a piRNA amplifier complex on transposon transcripts". Cell. 157 (7): 1698–1711. doi: 10.1016/j.cell.2014.05.018 . PMID   24910301.
  40. Mohn F, Handler D, Brennecke J (May 2015). "Noncoding RNA. piRNA-guided slicing specifies transcripts for Zucchini-dependent, phased piRNA biogenesis". Science. 348 (6236): 812–817. doi:10.1126/science.aaa1039. PMC   4988486 . PMID   25977553.
  41. Han BW, Wang W, Li C, Weng Z, Zamore PD (May 2015). "Noncoding RNA. piRNA-guided transposon cleavage initiates Zucchini-dependent, phased piRNA production". Science. 348 (6236): 817–821. doi:10.1126/science.aaa1264. PMC   4545291 . PMID   25977554.
  42. Le Thomas A, Rogers AK, Webster A, Marinov GK, Liao SE, Perkins EM, Hur JK, Aravin AA, Tóth KF (February 2013). "Piwi induces piRNA-guided transcriptional silencing and establishment of a repressive chromatin state". Genes & Development. 27 (4): 390–399. doi:10.1101/gad.209841.112. PMC   3589556 . PMID   23392610.
  43. 1 2 Wang G, Reinke V (June 2008). "A C. elegans Piwi, PRG-1, regulates 21U-RNAs during spermatogenesis". Current Biology. 18 (12): 861–867. Bibcode:2008CBio...18..861W. doi:10.1016/j.cub.2008.05.009. PMC   2494713 . PMID   18501605.
  44. Ozata DM, Gainetdinov I, Zoch A, Phillip D, Zamore PD (2019). "PIWI-interacting RNAs: small RNAs with big functions" (PDF). Nature Reviews Genetics . 20 (2): 89–108. doi:10.1038/s41576-018-0073-3. PMID   30446728. S2CID   53565676.
  45. Wu Q, Luo Y, Lu R, Lau N, Lai EC, Li WX; et al. (2010). "Virus discovery by deep sequencing and assembly of virus-derived small silencing RNAs". Proc Natl Acad Sci U S A. 107 (4): 1606–11. Bibcode:2010PNAS..107.1606W. doi: 10.1073/pnas.0911353107 . PMC   2824396 . PMID   20080648.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  46. Petit M, Mongelli V, Frangeul L, Blanc H, Jiggins F, Saleh MC (2016). "piRNA pathway is not required for antiviral defense in Drosophila melanogaster". Proc Natl Acad Sci U S A. 113 (29): E4218-27. Bibcode:2016PNAS..113E4218P. doi: 10.1073/pnas.1607952113 . PMC   4961201 . PMID   27357659.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  47. Campbell CL, Black WC, Hess AM, Foy BD (2008). "Comparative genomics of small RNA regulatory pathway components in vector mosquitoes". BMC Genomics. 9: 425. doi: 10.1186/1471-2164-9-425 . PMC   2566310 . PMID   18801182.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  48. 1 2 Varjak M, Maringer K, Watson M, Sreenu VB, Fredericks AC, Pondeville E; et al. (2017). "Aedes aegypti Piwi4 Is a Noncanonical PIWI Protein Involved in Antiviral Responses". mSphere. 2 (3). doi:10.1128/mSphere.00144-17. PMC   5415634 . PMID   28497119.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  49. Miesen P, Joosten J, van Rij RP (2016). "PIWIs Go Viral: Arbovirus-Derived piRNAs in Vector Mosquitoes". PLOS Pathog. 12 (12): e1006017. doi: 10.1371/journal.ppat.1006017 . PMC   5198996 . PMID   28033427.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  50. Parry R, Asgari S (2018). "Aedes Anphevirus: an Insect-Specific Virus Distributed Worldwide in Aedes aegypti Mosquitoes That Has Complex Interplays with Wolbachia and Dengue Virus Infection in Cells". J Virol. 92 (17). doi:10.1128/JVI.00224-18. PMC   6096813 . PMID   29950416.
  51. Parry R, Bishop C, De Hayr L, Asgari S (2019). "Density-dependent enhanced replication of a densovirus in Wolbachia-infected Aedes cells is associated with production of piRNAs and higher virus-derived siRNAs". Virology. 528: 89–100. doi: 10.1016/j.virol.2018.12.006 . PMID   30583288. S2CID   58572380.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  52. Kirino Y, Kim N, de Planell-Saguer M, Khandros E, Chiorean S, Klein PS, Rigoutsos I, Jongens TA, Mourelatos Z (May 2009). "Arginine methylation of Piwi proteins catalysed by dPRMT5 is required for Ago3 and Aub stability". Nat. Cell Biol. 11 (5): 652–8. doi:10.1038/ncb1872. PMC   2746449 . PMID   19377467.
  53. Anne J, Mechler BM (May 2005). "Valois, a component of the nuage and pole plasm, is involved in assembly of these structures, and binds to Tudor and the methyltransferase Capsuléen". Development. 132 (9): 2167–77. doi:10.1242/dev.01809. PMID   15800004. S2CID   6810484.
  54. Ro S, Park C, Jin J, Sanders KM, Yan W (December 2006). "A PCR-based method for detection and quantification of small RNAs". Biochemical and Biophysical Research Communications. 351 (3): 756–763. doi:10.1016/j.bbrc.2006.10.105. PMC   1934510 . PMID   17084816.
  55. Tang F, Hayashi K, Kaneda M, Lao K, Surani MA (May 2008). "A sensitive multiplex assay for piRNA expression". Biochemical and Biophysical Research Communications. 369 (4): 1190–1194. doi:10.1016/j.bbrc.2008.03.035. PMC   3855189 . PMID   18348866.
  56. Tosar JP, Rovira C, Cayota A (2018-01-22). "Non-coding RNA fragments account for the majority of annotated piRNAs expressed in somatic non-gonadal tissues". Communications Biology. 1 (1): 2. doi:10.1038/s42003-017-0001-7. PMC   6052916 . PMID   30271890.

Further reading