Hostname: page-component-745bb68f8f-f46jp Total loading time: 0 Render date: 2025-02-06T18:29:07.134Z Has data issue: false hasContentIssue false

MicroRNAs: Potentially important regulators for schistosome development and therapeutic targets against schistosomiasis

Published online by Cambridge University Press:  06 February 2012

GUOFENG CHENG*
Affiliation:
Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China, 200241
YOUXIN JIN
Affiliation:
State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, China, 200031; School of Life Sciences, Shanghai University, China, 200444
*
*Corresponding author: E-mail: Cheng_guofeng@yahoo.com
Rights & Permissions [Opens in a new window]

Summary

MicroRNAs (miRNAs) are small, endogenous non-coding RNA molecules that regulate gene expression post-transcriptionally by targeting the 3′ untranslated region (3′ UTR) of messenger RNAs. Since the discovery of the first miRNA in Caenorhabditis elegans, important regulatory roles for miRNAs in many key biological processes including development, cell proliferation, cell differentiation and apoptosis of many organisms have been described. Hundreds of miRNAs have been identified in various multicellular organisms and many are evolutionarily conserved. Schistosomes are multi-cellular eukaryotes with a complex life-cycle that require genes to be expressed and regulated precisely. Recently, miRNAs have been identified in two major schistosome species, Schistosoma japonicum and S. mansoni. These miRNAs are likely to play critical roles in schistosome development and gene regulation. Here, we review recent studies on schistosome miRNAs and discuss the potential roles of miRNAs in schistosome development and gene regulation. We also summarize the current status for targeting miRNAs and the potential of this approach for therapy against schistosomiasis.

Type
Research Article
Copyright
Copyright © Cambridge University Press 2012

INTRODUCTION

MicroRNAs (miRNAs) are a class of small non-coding RNAs (18–25 nucleotides in length) that are generated from endogenous transcripts that form hairpins (Kim, Reference Kim2005). miRNAs function as guide molecules for post-transcriptional gene regulation by base-pairing to their mRNAs targets (Bartel, Reference Bartel2004). Binding of miRNA to a target mRNA typically leads to translation repression and/or exonucleolytic mRNA decay. The first miRNA was discovered in the nematode Caenorhabditis elegans through genetic screens (Lee et al. Reference Lee, Feinbaum and Ambros1993; Reinhart et al. Reference Reinhart, Slack, Basson, Pasquinelli, Bettinger, Rougvie, Horvitz and Ruvkun2000). Since then many eukaryotic miRNA sequences and small non-coding RNAs have been identified and linked to a wide range of biological functions such as development, cell proliferation, metabolism, signal transduction and anti-viral defence. To date, over fifteen thousand miRNAs have been identified in over 140 species (http://www.mirbase.org, miRBase: release 17). Up to 30% of all human genes have been predicted to be regulated by miRNAs (Berezikov et al. Reference Berezikov, Guryev, van de Belt, Wienholds, Plasterk and Cuppen2005; Lewis et al. Reference Lewis, Burge and Bartel2005), suggesting that miRNAs serve a key regulatory role in the control gene expression.

Schistosomiasis is an important disease that afflicts ∼200 million people in 76 countries worldwide (Savioli et al. Reference Savioli, Stansfield, Bundy, Mitchell, Bhatia, Engels, Montresor, Neira and Shein2002; Hotez et al. Reference Hotez, Brindley, Bethony, King, Pearce and Jacobson2008). It is caused by parasitic worms of the genus Schistosoma, the most prevalent species being S. mansoni, S. japonicum and S. haematobium. No successful vaccine is currently available for this disease. Praziquantel is the only and most extensively used drug for treatment, but it is ineffective against young worms (Doenhoff and Pica-Mattoccia, Reference Doenhoff and Pica-Mattoccia2006) and there are concerns that drug resistance to praziquantel may be developing (Ismail et al. Reference Ismail, Botros, Metwally, William, Farghally, Tao, Day and Bennett1999). Consequently, there is an urgent need to identify novel drug targets and/or develop alternative therapeutic strategies.

Recently, miRNAs have been identified in schistosomes including both S. japonicum and S. mansoni. Here, we review the studies identifying schistosome miRNAs and their potential functions in schistosome development and gene regulation. We also summarize the current strategies for targeting miRNAs and the potential of this approach for schistosomiasis treatment or control.

miRNA BIOGENESIS

Primary miRNAs (pri-miRNAs) are transcribed by RNA polymerase II or III (Cai et al. Reference Cai, Hagedorn and Cullen2004; Borchert et al. Reference Borchert, Lanier and Davidson2006). The pri-miRNA is then processed by a nuclear microprocessor complex, composed of Drosha (a RNase III enzyme) and DGCR8 (DiGeorge critical region 8, also known as Pasha in Drosophila melanogaster and C. elegans), into a precursor-miRNA (pre-miRNA) that is a stem-loop hairpin precursor (Lee et al. Reference Lee, Ahn, Han, Choi, Kim, Yim, Lee, Provost, Radmark, Kim and Kim2003, Reference Lee, Kim, Han, Yeom, Lee, Baek and Kim2004). The pre-miRNA is then exported from the nucleus to the cytoplasm through exportin-5 in a Ran-GTP- and dsRNA-dependent manner, where it is further processed into a short duplex of 20–24nts by Dicer (another RNase III enzyme). Subsequently, a single strand from this duplex is preferentially loaded into the RNA Induced Silencing Complex (RISC) whose core binding protein is a member of the Argonaute protein family (Flynt and Lai, Reference Flynt and Lai2008). The miRNA/RISC binds directly to the 3′ UTR, 5′UTR or the open reading frame of the mRNAs through partial or complete complementary pairing between mRNA and miRNA sequences, typically resulting in mRNA degradation and/or protein translation inhibition (Moretti et al. Reference Moretti, Thermann and Hentze2010; Humphreys et al. Reference Humphreys, Westman, Martin and Preiss2005; Filipowicz et al. Reference Filipowicz, Bhattacharyya and Sonenberg2008; Qin et al. Reference Qin, Shi, Zhao, Yao, Jin, Ma and Jin2010). Recently, miRNAs have also been shown to be involved in translational activation (Filipowicz et al. Reference Filipowicz, Bhattacharyya and Sonenberg2008) and heterochromatin formation (Benetti et al. Reference Benetti, Gonzalo, Jaco, Munoz, Gonzalez, Schoeftner, Murchison, Andl, Chen, Klatt, Li, Serrano, Millar, Hannon and Blasco2008; Kim et al. Reference Kim, Saetrom, Snove and Rossi2008). Through these actions, miRNAs regulate gene expression during development, differentiation, proliferation, cell apoptosis and death, and metabolism in many organisms (Bushati and Cohen, Reference Bushati and Cohen2007). However, not all miRNAs are processed by this so-called canonical biogenesis pathway. Alternatively, some miRNAs are spliced and debranched from short intronic hairpins that bypass cleavage of Drosha, but nuclear export and further processing are common with the canonical miRNA pathway (Okamura et al. Reference Okamura, Hagen, Duan, Tyler and Lai2007; Ruby et al. Reference Ruby, Jan and Bartel2007).

Most miRNAs are under the control of developmental and/or tissue-specific signals and precise regulation of miRNA expression is crucial to maintain normal cellular functions. miRNA expression can be regulated both at the transcriptional level and at the post-transcriptional level. Many cellular transcription factors including c-Myc, p53, and E2F have been described to regulate miRNA transcription (He et al. Reference He, He, Lim, de Stanchina, Xuan, Liang, Xue, Zender, Magnus, Ridzon, Jackson, Linsley, Chen, Lowe, Cleary and Hannon2007; Woods et al. Reference Woods, Thomson and Hammond2007). Protein factors that interact with Drosha, Dicer and the terminal loop of pri- or pre-miRNAs can also regulate miRNA biogenesis. For example, p68/p72 enhances or represses maturation of a subset of miRNAs (94 out of 266 miRNAs investigated; Fukuda et al. Reference Fukuda, Yamagata, Fujiyama, Matsumoto, Koshida, Yoshimura, Mihara, Naitou, Endoh, Nakamura, Akimoto, Yamamoto, Katagiri, Foulds, Takezawa, Kitagawa, Takeyama, O'Malley and Kato2007). The KH-type splicing regulatory protein (KSRP) (Trabucchi et al. Reference Trabucchi, Briata, Garcia-Mayoral, Haase, Filipowicz, Ramos, Gherzi and Rosenfeld2009) and Lin28 (Viswanathan et al. Reference Viswanathan, Daley and Gregory2008), the specific interactors for miRNA precursor loops between RNA-binding proteins and cis-regulatory sequences, regulate the biogenesis of a subset of miRNAs.

SCHISTOSOME miRNA PATHWAY AND miRNA DISCOVERY

Schistosome miRNA pathway

The miRNA pathway consists of a conserved core of proteins and enzymes found in human, C. elegans, Drosophila melanogaster and Arabidopsis thaliana and known to function in the recognition and processing of miRNAs. Based on the most up-to-date model of miRNA biogenesis and regulated processing, bioinformatic analyses suggest that the schistosome genome encodes most of the known core factors and enzymes involved in miRNA biogenesis (Table 1). In S. mansoni, 13 putative proteins homologous with the known enzymes involved in the miRNA pathway in other organisms, including Dicer, Drosha, Ago and exportin-5, have been predicted using comparative genomics (Gomes et al. Reference Gomes, Cabral, Jannotti-Passos, Carvalho, Rodrigues, Baba and Sa2009). Since Ago proteins are the primary mediators of miRNA function, we identified three Ago proteins in S. japonicum and examined their expression profiles during schistosome development. Both S. japonicum eggs and miracidia showed relatively high expression of all three Ago proteins (Chen et al. Reference Chen, Yang, Guo, Peng, Liu, Li, Lin and Cheng2010b), a result suggesting that Ago proteins and/or their coordinated miRNAs may regulate gene expression and then control schistosome development and other biological processes (Chen et al. Reference Chen, Guo, Yang, Liu, Lin, Li and Cheng2010a, Reference Chen, Yang, Guo, Peng, Liu, Li, Lin and Chengb; Yang et al. Reference Yang, Guo, Chen, Lin, Liu and Cheng2010).

Table 1. Putative proteins involved in miRNA biogenesis and its regulation in schistosomes

* These putative proteins were also reported by Gomes (Gomes et al. Reference Gomes, Cabral, Jannotti-Passos, Carvalho, Rodrigues, Baba and Sa2009).

Schistosome miRNA discovery

To date, several groups have carried out the studies to identify miRNAs in schistosomes, particularly in S. japonicum (Table 2), and to provide some insight into the role of miRNAs in schistosome development.

Table 2. Summary of miRNA discovery in schistosomes

* A, Adult worms; C, Cercaria; S, Schistosomula.

To the best of our knowledge, Xue and co-workers first reported the identification of miRNAs in S. japonicum (Xue et al. Reference Xue, Sun, Zhang, Wang, Huang and Pan2008). They identified five miRNAs including Sja-let-7, Sja-miR-71, Sja-bantam, Sja-miR-125 and Sja-miR-new1 in S. japonicum by direct cloning. They reported that four out of five miRNAs were shown to be conserved in other organisms and that the miRNA expression profiles were highly stage-specific (Xue et al. Reference Xue, Sun, Zhang, Wang, Huang and Pan2008). In a more comprehensive study, Wang and co-workers used a high-throughput sequencing technology, Illumina Solexa, to characterize small RNAs populations in S. japonicum schistosomula, the early stage in the vertebrate host. They identified 20 evolutionarily conserved miRNAs and 16 schistosome-specific miRNAs (Wang et al. Reference Wang, Xue, Sun, Luo, Xu, Jiang, Zhang and Pan2010). Using a similar approach, Huang et al. (Reference Huang, Hao, Chen, Hu, Yan, Liu and Han2009) reported 176 species-specific miRNAs in immature schistosomula and mature, paired adult S. japonicum while Hao and co-workers identified 16 evolutionarily conserved miRNAs and 22 schistosome-specific miRNAs (Hao et al. Reference Hao, Cai, Jiang, Wang and Chen2010) in the same organism. In S. mansoni, Simoes et al. (Reference Simoes, Lee, Djikeng, Cerqueira, Zerlotini, da Silva-Pereira, Dalby, LoVerde, El-Sayed and Oliveira2011) identified 211 novel miRNA candidates by sequencing small RNA cDNA libraries from adult worm pairs and presented data supporting stage-regulated expression patterns for some of the miRNAs.

Although the Illumina deep sequencing has been applied for schistosome miRNA discovery in several studies, the number of miRNAs identified showed significant variation between these studies (Table 2). Differing starting material and procedures used for RNA isolation, library construction and/or sequencing can affect the number of miRNAs identified. The criteria used to define a miRNA in these studies were different and less stringent than those currently used for high-confidence miRNA annotation (Meyers et al. Reference Meyers, Axtell, Bartel, Bartel, Baulcombe, Bowman, Cao, Carrington, Chen, Green, Griffiths-Jones, Jacobsen, Mallory, Martienssen, Poethig, Qi, Vaucheret, Voinnet, Watanabe, Weigel and Zhu2008; Berezikov et al. Reference Berezikov, Liu, Flynt, Hodges, Rooks, Hannon and Lai2010; Chiang et al. Reference Chiang, Schoenfeld, Ruby, Auyeung, Spies, Baek, Johnston, Russ, Luo, Babiarz, Blelloch, Schroth, Nusbaum and Bartel2010). For high-confidence miRNA annotation, apart from the requirement that the miRNA should be derived from a unique and relatively stable genomic hairpin predicted by bioinformatics, the following criteria should be also jointly considered: (1) miRNAs have multiple cloned 21-24nt reads with relatively fixed 5′ ends; (2) the miRNA* (an miRNA* denotes the small RNA processed from the hairpin arm opposite the mature miRNA) is also presented in the reads; (3) the miRNA and miRNA* form a duplex with two nucleotide, 3′ overhangs; (4) with sufficient sequencing, it is also possible to present other byproducts of miRNA biogenesis such as terminal loops or species flanking the pre-miRNA hairpin. The defined miRNAs in schistosomes will be narrowed significantly when these combined criteria are applied. Nevertheless, these studies have still provided substantial evidence to support the view that miRNAs serve as critical regulators in the regulation of gene expression in schistosomes and also imply miRNA roles in schistosome development.

EVOLUTIONARILY CONSERVED miRNAS IN SCHISTOSOMES

Many miRNAs from bilateral animals are phylogenetically conserved; 55% of C. elegans miRNAs have homologues in humans. This suggests that miRNAs have played important roles throughout animal evolution (Pasquinelli et al. Reference Pasquinelli, Reinhart, Slack, Martindale, Kuroda, Maller, Hayward, Ball, Degnan, Muller, Spring, Srinivasan, Fishman, Finnerty, Corbo, Levine, Leahy, Davidson and Ruvkun2000; Ibanez-Ventoso et al. Reference Ibanez-Ventoso, Vora and Driscoll2008). Some conserved miRNAs are involved in critical biological processes such as cell differentiation and proliferation, apoptosis, metabolism and development (Reinhart et al. Reference Reinhart, Slack, Basson, Pasquinelli, Bettinger, Rougvie, Horvitz and Ruvkun2000; Johnson et al. Reference Johnson, Grosshans, Shingara, Byrom, Jarvis, Cheng, Labourier, Reinert, Brown and Slack2005; Chen et al. Reference Chen, Mandel, Thomson, Wu, Callis, Hammond, Conlon and Wang2006; Caygill and Johnston, Reference Caygill and Johnston2008). Furthermore, it has been demonstrated that an evolutionarily conserved miRNA coevolves with its target mRNA, retaining a similar development or physiological role in diverse taxa (Lee and Ambros, Reference Lee and Ambros2001; Takane et al. Reference Takane, Fujishima, Watanabe, Sato, Saito, Tomita and Kanai2010).

Based on the conserved miRNAs reported by individual studies, we selected S. japonicum miRNAs that have been identified in several independent studies and examined their evolutionary conservation in humans (Homo sapiens), Mus musculus, D. melanogaster, C. elegans and a member of the same phylum (Platyhleminthes) as schistosomes, the fresh-water planarian Schmidtea mediterranea. Briefly, S. japonicum miRNAs were searched against the miRbase by using BLASTN (http://www.mirbase.org) at the default setting and then the pairs were subjected to manual checks. As summarized in Table 3, thirteen S. japonicum miRNAs have corresponding homologues in at least three species out of five investigated (Table 3). Five miRNAs including miR-1b, miR-124, miR-8/200b, miR-190 and miR-7 are conserved among all five species (Table 3).

Table 3. Evolutionarily conserved miRNAs in S. japonicum

Some of these conserved miRNAs have been functionally characterized in mammalian cells, C. elegans or Drosophila and were also shown to play important roles in regulating cell differentiation, cell survival, apoptosis and proliferation (Table 4). Briefly, miR-1 is a very widely conserved miRNA with specific expression in muscle and heart (Lee and Ambros, Reference Lee and Ambros2001; Lagos-Quintana et al. Reference Lagos-Quintana, Rauhut, Yalcin, Meyer, Lendeckel and Tuschl2002; Kwon et al. Reference Kwon, Han, Olson and Srivastava2005) and such diverse targets as heat shock proteins, twinfilin-1, transgelin 2, et al. (Yang et al. Reference Yang, Lin, Xiao, Lu, Luo, Li, Zhang, Xu, Bai, Wang, Chen and Wang2007; Simon et al. Reference Simon, Madison, Conery, Thompson-Peer, Soskis, Ruvkun, Kaplan and Kim2008; Yan et al. Reference Yan, Dong Xda, Chen, Wang, Lu, Wang, Qu and Tu2009; Li et al. Reference Li, Song, Zou, Wang, Kremneva, Li, Zhu, Sun, Lappalainen, Yuan, Qin and Jing2010; Nohata et al. Reference Nohata, Sone, Hanazawa, Fuse, Kikkawa, Yoshino, Chiyomaru, Kawakami, Enokida, Nakagawa, Shozu, Okamoto and Seki2011; Yoshino et al. Reference Yoshino, Chiyomaru, Enokida, Kawakami, Tatarano, Nishiyama, Nohata, Seki and Nakagawa2011) (Table 4). miR-1 targets transcription factors such as Kruppel-like factor 4 (KLF4) (Xie et al. Reference Xie, Huang, Sun, Guo, Hamblin, Ritchie, Garcia-Barrio, Zhang and Chen2011) and Paired-box transcription factor (Chen et al. Reference Chen, Tao, Li, Deng, Yan, Xiao and Wang2010c). Most interestingly, a recent study indicated that miR-1 can interact with insulin-like growth factor-1 (IGF-1) and IGF-1 receptor and this activation of the IGF-1 signal pathway reciprocally regulates miR-1 expression (Elia et al. Reference Elia, Contu, Quintavalle, Varrone, Chimenti, Russo, Cimino, De Marinis, Frustaci, Catalucci and Condorelli2009). In schistosomes, the insulin receptors have been characterized preliminarily in both S. mansoni (Khayath et al. Reference Khayath, Vicogne, Ahier, BenYounes, Konrad, Trolet, Viscogliosi, Brehm and Dissous2007) and S. japonicum (You et al. Reference You, Zhang, Jones, Gobert, Mulvenna, Rees, Spanevello, Blair, Duke, Brehm and McManus2010). Both species of schistosomes have two distinct insulin receptor homologues with different expression patterns. The two receptors can bind to human pro-insulin with high affinity in yeast two-hybrid assays (Khayath et al. Reference Khayath, Vicogne, Ahier, BenYounes, Konrad, Trolet, Viscogliosi, Brehm and Dissous2007; You et al. Reference You, Zhang, Jones, Gobert, Mulvenna, Rees, Spanevello, Blair, Duke, Brehm and McManus2010) and there is a link between the functions of the insulin receptor and worm glucose consumption (You et al. Reference You, Zhang, Jones, Gobert, Mulvenna, Rees, Spanevello, Blair, Duke, Brehm and McManus2010). Additionally, the IGF-1 signal transduction cascade is also subject to miRNA-145 regulation in mammalian cells (La Rocca et al. Reference La Rocca, Badin, Shi, Xu, Deangelis, Sepp-Lorenzinoi and Baserga2009). It will be interesting to investigate whether schistosome miR-1 regulates IGF-1 receptor signaling and the role it plays in parasite development, growth and host-parasite interactions. Next, miR-124 is also tissue specific being found in the nervous system of all animals studied to date (Lagos-Quintana et al. Reference Lagos-Quintana, Rauhut, Yalcin, Meyer, Lendeckel and Tuschl2002; Nelson et al. Reference Nelson, Baldwin, Kloosterman, Kauppinen, Plasterk and Mourelatos2006) and plays a vital role in neuronal development (Lim et al. Reference Lim, Lau, Garrett-Engele, Grimson, Schelter, Castle, Bartel, Linsley and Johnson2005; Krichevsky et al. Reference Krichevsky, Sonntag, Isacson and Kosik2006; Kapsimali et al. Reference Kapsimali, Kloosterman, de Bruijn, Rosa, Plasterk and Wilson2007; Makeyev et al. Reference Makeyev, Zhang, Carrasco and Maniatis2007) (Table 4). In brain tumours, miR-124 can modulate medulloblastoma cell growth by targeting cyclin dependent kinase 6 (CDK6) (Pierson et al. Reference Pierson, Hostager, Fan and Vibhakar2008) and solute carrier family 16 (SLC16A1) (Li et al. 2009). Although genome studies suggest that the schistosome nervous and sensory system may be central to parasite migration and development (Zhou et al. Reference Zhou, Zheng, Chen, Zhang, Wang, Guo, Huang, Zhang, Huang, Jin, Dou, Hasegawa, Wang, Zhang, Zhou, Tao, Cao, Li, Vinar, Brejova, Brown, Li, Miller, Blair, Zhong, Chen, Liu, Hu, Wang, Zhang, Song, Chen, Xu, Xu, Ju, Huang, Brindley, McManus, Feng, Han, Lu, Ren, Wang, Gu, Kang, Chen, Chen, Chen, Wang, Yan, Wang, Lv, Jin, Wang, Pu, Zhang, Zhang, Hu, Zhu, Wang, Yu, Wang, Yang, Ning, Beriman, Wei, Ruan, Zhao, Wang, Liu, Zhou, Wang, Lu, Zheng, Brindley, McManus, Blair, Zhang, Zhong, Wang, Han, Chen, Wang, Han and Chen2009), the molecular basis is poorly characterized. Moreover, miR-8/200b has been recognized as a critical mediator for several well characterized developmental regulatory networks such as Wnt, Notch, and TGF-beta in Drosophila or mammalian cells (Table 4). In addition, these signaling pathways have also been implicated in critical roles in schistosome development by the S. japonicum genome study (Zhou et al. Reference Zhou, Zheng, Chen, Zhang, Wang, Guo, Huang, Zhang, Huang, Jin, Dou, Hasegawa, Wang, Zhang, Zhou, Tao, Cao, Li, Vinar, Brejova, Brown, Li, Miller, Blair, Zhong, Chen, Liu, Hu, Wang, Zhang, Song, Chen, Xu, Xu, Ju, Huang, Brindley, McManus, Feng, Han, Lu, Ren, Wang, Gu, Kang, Chen, Chen, Chen, Wang, Yan, Wang, Lv, Jin, Wang, Pu, Zhang, Zhang, Hu, Zhu, Wang, Yu, Wang, Yang, Ning, Beriman, Wei, Ruan, Zhao, Wang, Liu, Zhou, Wang, Lu, Zheng, Brindley, McManus, Blair, Zhang, Zhong, Wang, Han, Chen, Wang, Han and Chen2009). Finally, miR-7 has been shown to interact with EGF (epidermal growth factor) and IGF-1 receptors in mammalian cells (Kefas et al. Reference Kefas, Godlewski, Comeau, Li, Abounader, Hawkinson, Lee, Fine, Chiocca, Lawler and Purow2008; Webster et al. Reference Webster, Giles, Price, Zhang, Mattick and Leedman2009; Jiang et al. Reference Jiang, Liu, Chen, Jin, Heidbreder, Kolokythas, Wang, Dai and Zhou2010). Both these receptor mediated signaling pathways typically promote cell survival, growth and differentiation via the activation of several integrated signaling pathways (Taguchi and White, Reference Taguchi and White2008; Hynes and MacDonald, Reference Hynes and MacDonald2009). In S. mansoni, the growth factor receptor (SER) was shown to be present predominantly in muscle tissue (Shoemaker et al. Reference Shoemaker, Ramachandran, Landa, dos Reis and Stein1992), implying a role in muscle development and function. Furthermore, the schistosome SER can bind human EGF with high affinity when expressed in heterologous vertebrate cells and this binding can lead to SER phosphorylation in S. mansoni membranes as well as activation of a conserved Ras/extracelluar regulated kinase dependent signaling pathway, suggesting the important role of EGF-receptor in the parasite development (Vicogne et al. Reference Vicogne, Cailliau, Tulasne, Browaeys, Yan, Fafeur, Vilain, Legrand, Trolet and Dissous2004; Dissous et al. Reference Dissous, Khayath, Vicogne and Capron2006; You et al. Reference You, Gobert, Jones, Zhang and McManus2011). Overall, these conserved miRNAs appear to contribute to the regulation of several signaling pathways such as IGF (miR-1/miR-7), WNT (miR-8/200b), Notch (miR-8/200b), TGF-beta (miR-8/200b), and EGF (miR-7) in other organisms and these pathways have been shown to be potentially important for regulating schistosome development.

Table 4. Summary of the targets of evolutionarily conserved miRNAs for schistosomes

POSSIBLE ROLES OF miRNAS IN SCHISTOSOME DEVELOPMENT

Molecular basis of schistosome development

Schistosomes have a complex life-cycle with distinct developmental stages in the molluscan and vertebrate hosts that exhibit significant morphological and physiological differences. Firstly, transcriptome analysis has demonstrated that the S. japonicum genome encodes mammalian-like receptors for insulin, progesterone, cytokines and neuropeptides, suggesting that host hormones, or endogenous parasite homologues, could orchestrate schistosome development and maturation (Hu et al. 2003). Next, the S. japonicum genome study indicated the presence of sequences that encode some growth factors, receptors and essential components for well-characterized signaling pathways such as Wnt, notch, hedgehog, IGF, EGF and transforming growth factor β (TGF-β; Zhou et al. Reference Zhou, Zheng, Chen, Zhang, Wang, Guo, Huang, Zhang, Huang, Jin, Dou, Hasegawa, Wang, Zhang, Zhou, Tao, Cao, Li, Vinar, Brejova, Brown, Li, Miller, Blair, Zhong, Chen, Liu, Hu, Wang, Zhang, Song, Chen, Xu, Xu, Ju, Huang, Brindley, McManus, Feng, Han, Lu, Ren, Wang, Gu, Kang, Chen, Chen, Chen, Wang, Yan, Wang, Lv, Jin, Wang, Pu, Zhang, Zhang, Hu, Zhu, Wang, Yu, Wang, Yang, Ning, Beriman, Wei, Ruan, Zhao, Wang, Liu, Zhou, Wang, Lu, Zheng, Brindley, McManus, Blair, Zhang, Zhong, Wang, Han, Chen, Wang, Han and Chen2009). Among these putative pathways, TGF-β is the most well characterized one in schistosomes so far. Several studies indicated the multiple roles of TGF-β throughout schistosome development. These include the interaction between the male and female schistosome and between parasite and host as well as development of vitelline cells in female worms whose genes are regulated by a stimulus from the male schistosome and embryogenesis of the egg (Osman et al. Reference Osman, Niles, Verjovski-Almeida and LoVerde2006; Freitas et al. Reference Freitas, Jung and Pearce2007; Loverde et al. Reference Loverde, Osman and Hinck2007). These obeservations suggest that schistosome parasites have co-evolved an intricate relationship with their hosts as well as a novel interplay between the adult male and female parasites.

Although direct evidence for miRNA mediated TGF-β signaling in schistosomes remains to be experimentally clarified, several mammalian miRNAs such as miR-141 (Hu et al. 2010), miR-244 (Yao et al. Reference Yao, Yin, Lian, Tian, Liu, Li and Sun2010), and miR-23b (Yuan et al. Reference Yuan, Dong, Shi, Zhou, Zhao, Miao and Jiao2011) are involving in TGF-β signal pathways by targeting Smad proteins (intracellular proteins that transduce extracellular signals from TGF-β ligands to the nucleus) in mammalian cells or organs to mediate cell proliferation or organ regeneration. Additionally, Smad proteins have also been documented to control Drosha-mediated miRNA maturation as reported by Davis et al. (Reference Davis, Hilyard, Lagna and Hata2008). In this study, they demonstrated that TGF-β signaling promotes the expression of mature miR-21 by increasing the processing of primary transcripts of miR-21 into precursor miR-21 by the Drosha complex (Davis et al. Reference Davis, Hilyard, Lagna and Hata2008). Together these studies suggest an intricate relationship between TGF-β signaling and miRNAs to regulate embryonic development and maintaining tissue homeostasis in higher organisms.

Putative miRNA:mRNA target pairs potentially involved in schistosome development

As described above, phylogenetically conserved miRNAs regulate a number of signal pathways such as Wnt, VEGF, and TGF-β (Kennell et al. Reference Kennell, Gerin, MacDougald and Cadigan2008; Elia et al. Reference Elia, Contu, Quintavalle, Varrone, Chimenti, Russo, Cimino, De Marinis, Frustaci, Catalucci and Condorelli2009; Webster et al. Reference Webster, Giles, Price, Zhang, Mattick and Leedman2009) in other organisms. These pathways are likely to play critical roles in schistosome development (Osman et al. Reference Osman, Niles, Verjovski-Almeida and LoVerde2006; Zhou et al. Reference Zhou, Zheng, Chen, Zhang, Wang, Guo, Huang, Zhang, Huang, Jin, Dou, Hasegawa, Wang, Zhang, Zhou, Tao, Cao, Li, Vinar, Brejova, Brown, Li, Miller, Blair, Zhong, Chen, Liu, Hu, Wang, Zhang, Song, Chen, Xu, Xu, Ju, Huang, Brindley, McManus, Feng, Han, Lu, Ren, Wang, Gu, Kang, Chen, Chen, Chen, Wang, Yan, Wang, Lv, Jin, Wang, Pu, Zhang, Zhang, Hu, Zhu, Wang, Yu, Wang, Yang, Ning, Beriman, Wei, Ruan, Zhao, Wang, Liu, Zhou, Wang, Lu, Zheng, Brindley, McManus, Blair, Zhang, Zhong, Wang, Han, Chen, Wang, Han and Chen2009). Although these evolutionarily conserved miRNAs and the corresponding mRNA targets coexist in schistosomes, it remains unknown whether the miRNAs regulate similar mRNAs.

Five highly conserved miRNAs (miR-1, miR-124, miR-8/200b, miR-195, and miR-7) are predicted to interact with possible phylogenetically conserved mRNA targets in schistosomes based on analysis of miRNA:mRNA pairs predicted using RNAhybrid (http://bibiserv.techfak.uni-bielefeld.de/rnahybrid/) and PITA (http://genie.weizmann.ac.il/pubs/mir07/mir07_prediction.html). Based on predicted potential targets, signaling pathways such as Wnt, VEGF and TGF-β and such important biological processes as cell cycle regulation and actin reorganization in S. japoncum may be regulated by the evolutionarily conserved miRNAs (Table 5). These predicted interactions and their roles in schistosome development suggest interesting avenues for future experimentation.

Table 5. List of some putative evolutionarily conserved miRNA:mRNA target pairs in S. japonicum

THERAPEUTIC POTENTIAL FOR SCHISTOSOMIASIS CONTROL BY TARGETING miRNAS

Given their essential roles in many biological functions, miRNAs have considerable potential anti-pathogen therapy (Wurdinger and Costa, Reference Wurdinger and Costa2007). To date, several strategies have been used to silence miRNAs in either mammalian cells or model organisms such as Drosophila and C. elegans or even in higher organisms such as mice and primate.

Firstly, antisense oligonucleotides (ASOs) complementary to the target miRNA mediate miRNA silencing. ASOs could be single strand RNA (Krutzfeldt et al. Reference Krutzfeldt, Rajewsky, Braich, Rajeev, Tuschl, Manoharan and Stoffel2005) or DNA (Esau et al. Reference Esau, Davis, Murray, Yu, Pandey, Pear, Watts, Booten, Graham, McKay, Subramaniam, Propp, Lollo, Freier, Bennett, Bhanot and Monia2006) analogues, or locked nuclear acids (LNA) (Elmen et al. Reference Elmen, Lindow, Schutz, Lawrence, Petri, Obad, Lindholm, Hedtjarn, Hansen, Berger, Gullans, Kearney, Sarnow, Straarup and Kauppinen2008) produced by chemical synthesis. The first study using ASOs for miRNA silencing was reported by Boutla et al. in 2003. In this study, the authors injected the DNA oligonucleotides complementary to 11 miRNAs into Drosophila embryos and then observed a variety of developmental defects, implying that ASO-mediated miRNA inhibition could interfere with Drosophila development. Then, chemically modified- or cholesterol-conjugated ASOs have also been used for miRNA silencing due to their relatively long stability and the high efficiency of delivery. For example, Hutvagner and co-workers demonstrated that injection of the 2′-O-methyl modified oligonucleotide complementary to the miRNA let-7 in C. elegans caused L4 larval moult defects and the inappropriate production of the larval cuticle at the adult stage (Hutvagner et al. Reference Hutvagner, Simard, Mello and Zamore2004). Then, Krützfeldt and co-workers injected chemically modified, cholesterol-conjugated single-stranded RNA analogues (termed antagomirs) complementary to miRNAs (miR-16, miR-122, miR-192 and miR-194) in mice and subsequently observed a marked reduction of corresponding miRNA levels in many organs and tissues of mice such as liver, lung, kidney, heart, intestine, fat, skin, bone marrow, muscle and ovaries. Notably, the silencing of endogenous miRNAs by these antagomirs is specific, efficient and long-lasting (Krutzfeldt et al. Reference Krutzfeldt, Rajewsky, Braich, Rajeev, Tuschl, Manoharan and Stoffel2005). To move this therapeutic potential toward human applications, Elmén et al. (Reference Elmen, Lindow, Schutz, Lawrence, Petri, Obad, Lindholm, Hedtjarn, Hansen, Berger, Gullans, Kearney, Sarnow, Straarup and Kauppinen2008) designed an unconjugated, PBS-formulated locked nucleic acid-modified oligonucleotide (LNA-antimiR) and injected this LNA-antimiR (complementary to miR-122; 3 or 10 mg/kg) into a non-human primate (African green monkeys) intravenously. They observed that the injected LNA-antimiR was taken up in the cytoplasm of the hepatocytes and formed stable heteroduplexes with miR-122, leading to the depletion of mature miR-122 and dose-dependent lowering of plasma cholesterol. Since mammals including humans and mice can be final hosts for schistosomes, targeting of schistosome-specific miRNAs by using these antagomirs may offer a potential for schistosomiasis treatment. Through the ASO-based gene modulation, several therapeutic molecules are in clinical trials (Goodchild, Reference Goodchild2004; Chi et al. Reference Chi, Eisenhauer, Fazli, Jones, Goldenberg, Powers, Tu and Gleave2005), but possible toxicity and off-target effects will need to be addressed (Esau, Reference Esau2008). Alternatively, morpholino oligos, which are also synthetic nucleic acid molecules, can block miRNA activity and also may eliminate off-target effects. Kloosterman and co-workers firstly employed morpholino oligos to knock down 13 miRNAs conserved in zebrafish and subsequently demonstrated that knockdown of miR-375 affected the morphology of the pancreatic islet (Kloosterman et al. Reference Kloosterman, Lagendijk, Ketting, Moulton and Plasterk2007).

Vector-based miRNA inhibition is another strategy being tested for in vivo miRNA silencing. Expression cassettes for short antisense RNAs can be engineered into lentiviral or adenoviral vectors to give stable or transient expression in cell lines or whole animals (Poy et al. Reference Poy, Eliasson, Krutzfeldt, Kuwajima, Ma, Macdonald, Pfeffer, Tuschl, Rajewsky, Rorsman and Stoffel2004; Scherr et al. Reference Scherr, Venturini, Battmer, Schaller-Schoenitz, Schaefer, Dallmann, Ganser and Eder2007). Vector-based miRNA inhibition offers several advantages compared to chemically synthesized antagomirs, including longevity, cost-effectiveness with the potential for site-specific potential by engineering suitable promoters.

miRNA inhibition can also be achieved by targeting essential factors involved in miRNA biogenesis such as Drosha, DGCR8, Dicer and Ago using small interfering RNAs (siRNAs). However, the strategy results in globally reduced mature miRNAs, leading to severe off-target effects. Additionally, Ebert et al. (Reference Ebert, Neilson and Sharp2007) reported a competitive miRNA inhibitor, termed miRNA sponges which can specifically inhibit miRNAs with a complementary heptameric seed. However, further understanding of miRNA biogenesis and the functional mechanisms is critical for the effective optimization of miRNA silencing, be it through optimal design, chemical modification and/or delivery of the miRNA antagoniser.

The use of chemically synthesized siRNAs (Cheng et al. Reference Cheng, Fu, Lin, Shi, Zhou, Jin and Cai2009) or vector-based short hairpin RNA (Zhao et al. Reference Zhao, Lei, Liu, Zhu, Ren, Wang and Shen2008) can significantly inhibit endogenous mRNAs in several different stages such as schistosomula, adult worms and sporocysts (Boyle et al. Reference Boyle, Wu, Shoemaker and Yoshino2003). Targeting of the schistosome gynaecophoral canal protein in S. japonicum and hypoxanthine-guanine phosphoribosyltransferase in S. mansoni by siRNAs has been successfully applied in vivo leading to a significant worm burden reduction (Pereira et al. Reference Pereira, Pascoal, Marchesini, Maia, Magalhaes, Zanotti-Magalhaes and Lopes-Cendes2008; Cheng et al. Reference Cheng, Fu, Lin, Shi, Zhou, Jin and Cai2009). These preliminary studies suggest in vivo therapeutic gene modulation in schistosomes by delivering small nuclear acids may have the potential for schistosomiasis treatment. Although siRNAs are not encoded in the genome of organisms unlike miRNAs which function as natural regulators of gene expression, most antagonisers for miRNA silencing are also small nuclear acids. Consequently, it is reasonable to suggest that the usage of ASOs, LNA or vector-based miRNA antisense targeting schistosome miRNAs may have the potential for schistosomiasis treatment.

CONCLUSIONS

miRNAs play a vital role in many biological processes. Understanding of miRNA functions in schistosomes may provide insights into schistosome development and growth as well as sexual maturation and may lead to the identification of novel therapeutic targets for schistosomiasis. Furthermore, targeting of schistosome-specific miRNAs by ASOs, LNA or miRNA mimics may have therapeutic potential for schistosomiasis control. For this to be realized, a deep understanding of miRNA biogenesis and miRNA functions in schistosomes and their hosts is critical to identify a therapeutic target and develop effective and efficient antagomirs for schistosomes.

FINANCIAL SUPPORT

The study was supported, in part or in whole, by National Natural Science Foundation of China (Grant No. 30901068), Science and Technology Commission of Shanghai Municipality of China (Grant No. 10PJ1412300), Shanghai Talent Developing Foundation of China (Grant No. 2009032) and Basic Foundation for Scientific Research of State-level Public Welfare Institutes of China (Grant No. 2009JB08).

References

REFERENCES

Bartel, D. P. (2004). MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 281297.CrossRefGoogle ScholarPubMed
Benetti, R., Gonzalo, S., Jaco, I., Munoz, P., Gonzalez, S., Schoeftner, S., Murchison, E., Andl, T., Chen, T., Klatt, P., Li, E., Serrano, M., Millar, S., Hannon, G. and Blasco, M. A. (2008). A mammalian microRNA cluster controls DNA methylation and telomere recombination via Rbl2-dependent regulation of DNA methyltransferases. Nature Structural & Molecular Biology 15, 268279.CrossRefGoogle ScholarPubMed
Berezikov, E., Guryev, V., van de Belt, J., Wienholds, E., Plasterk, R. H. and Cuppen, E. (2005). Phylogenetic shadowing and computational identification of human microRNA genes. Cell 120, 2124.CrossRefGoogle ScholarPubMed
Berezikov, E., Liu, N., Flynt, A. S., Hodges, E., Rooks, M., Hannon, G. J. and Lai, E. C. (2010). Evolutionary flux of canonical microRNAs and mirtrons in Drosophila. Nature Genetics 42, 69; author reply 9–10.CrossRefGoogle ScholarPubMed
Borchert, G. M., Lanier, W. and Davidson, B. L. (2006). RNA polymerase III transcribes human microRNAs. Nature Structural & Molecular Biology 13, 10971101.CrossRefGoogle ScholarPubMed
Boutla, A., Delidakis, C. and Tabler, M. (2003). Developmental defects by antisense-mediated inactivation of micro-RNAs 2 and 13 in Drosophila and the identification of putative target genes. Nucleic Acids Research 31, 49734980.CrossRefGoogle ScholarPubMed
Boyle, J. P., Wu, X. J., Shoemaker, C. B. and Yoshino, T. P. (2003). Using RNA interference to manipulate endogenous gene expression in Schistosoma mansoni sporocysts. Molecular and Biochemical Parasitology 128, 205215.CrossRefGoogle ScholarPubMed
Bushati, N. and Cohen, S. M. (2007). microRNA functions. Annual Review of Cell and Developmental Biology 23, 175205.CrossRefGoogle ScholarPubMed
Cai, X., Hagedorn, C. H. and Cullen, B. R. (2004). Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs. RNA 10, 19571966.CrossRefGoogle ScholarPubMed
Cao, X., Pfaff, S. L. and Gage, F. H. (2007). A functional study of miR-124 in the developing neural tube. Genes & Development 21, 531536.CrossRefGoogle ScholarPubMed
Caygill, E. E. and Johnston, L. A. (2008). Temporal regulation of metamorphic processes in Drosophila by the let-7 and miR-125 heterochronic microRNAs. Current Biology 18, 943950.CrossRefGoogle ScholarPubMed
Chen, J., Guo, S. X., Yang, Y. P., Liu, J. M., Lin, J. J., Li, J. K. and Cheng, G. F. (2010 a). Cloning and preliminary analysis of a full-length cDNA encoding Argonaute 3 from Schistosoma japonicum. Chinese Journal Schistosomiasis Control 22, 310315.Google Scholar
Chen, J. F., Mandel, E. M., Thomson, J. M., Wu, Q., Callis, T. E., Hammond, S. M., Conlon, F. L. and Wang, D. Z. (2006). The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nature Genetics 38, 228233.CrossRefGoogle ScholarPubMed
Chen, J. F., Tao, Y., Li, J., Deng, Z., Yan, Z., Xiao, X. and Wang, D. Z. (2010 c). microRNA-1 and microRNA-206 regulate skeletal muscle satellite cell proliferation and differentiation by repressing Pax7. Journal of Cell Biology 190, 867879.CrossRefGoogle ScholarPubMed
Chen, J., Yang, Y., Guo, S., Peng, J., Liu, Z., Li, J., Lin, J. and Cheng, G. (2010 b). Molecular cloning and expression profiles of Argonaute proteins in Schistosoma japonicum. Parasitology Research 107, 889899.CrossRefGoogle ScholarPubMed
Cheng, G., Fu, Z., Lin, J., Shi, Y., Zhou, Y., Jin, Y. and Cai, Y. (2009). In vitro and in vivo evaluation of small interference RNA-mediated gynaecophoral canal protein silencing in Schistosoma japonicum. Journal of Gene Medicine 11, 412421.CrossRefGoogle ScholarPubMed
Chi, K. N., Eisenhauer, E., Fazli, L., Jones, E. C., Goldenberg, S. L., Powers, J., Tu, D. and Gleave, M. E. (2005). A phase I pharmacokinetic and pharmacodynamic study of OGX-011, a 2′-methoxyethyl antisense oligonucleotide to clusterin, in patients with localized prostate cancer. Journal of the National Cancer Institute 97, 12871296.CrossRefGoogle Scholar
Chiang, H. R., Schoenfeld, L. W., Ruby, J. G., Auyeung, V. C., Spies, N., Baek, D., Johnston, W. K., Russ, C., Luo, S., Babiarz, J. E., Blelloch, R., Schroth, G. P., Nusbaum, C. and Bartel, D. P. (2010). Mammalian microRNAs: experimental evaluation of novel and previously annotated genes. Genes & Development 24, 9921009.CrossRefGoogle ScholarPubMed
Davis, B. N., Hilyard, A. C., Lagna, G. and Hata, A. (2008). SMAD proteins control DROSHA-mediated microRNA maturation. Nature 454, 5661.CrossRefGoogle ScholarPubMed
Dissous, C., Khayath, N., Vicogne, J. and Capron, M. (2006). Growth factor receptors in helminth parasites: signalling and host-parasite relationships. FEBS Letters 580, 29682975.CrossRefGoogle ScholarPubMed
Doenhoff, M. J. and Pica-Mattoccia, L. (2006). Praziquantel for the treatment of schistosomiasis: its use for control in areas with endemic disease and prospects for drug resistance. Expert Review of Anti-Infective Therapy 4, 199210.CrossRefGoogle ScholarPubMed
Ebert, M. S., Neilson, J. R. and Sharp, P. A. (2007). MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nature Methods 4, 721726.CrossRefGoogle ScholarPubMed
Elia, L., Contu, R., Quintavalle, M., Varrone, F., Chimenti, C., Russo, M. A., Cimino, V., De Marinis, L., Frustaci, A., Catalucci, D. and Condorelli, G. (2009). Reciprocal regulation of microRNA-1 and insulin-like growth factor-1 signal transduction cascade in cardiac and skeletal muscle in physiological and pathological conditions. Circulation 120, 23772385.CrossRefGoogle ScholarPubMed
Elmen, J., Lindow, M., Schutz, S., Lawrence, M., Petri, A., Obad, S., Lindholm, M., Hedtjarn, M., Hansen, H. F., Berger, U., Gullans, S., Kearney, P., Sarnow, P., Straarup, E. M. and Kauppinen, S. (2008). LNA-mediated microRNA silencing in non-human primates. Nature 452, 896899.CrossRefGoogle ScholarPubMed
Esau, C. C. (2008). Inhibition of microRNA with antisense oligonucleotides. Methods 44, 5560.CrossRefGoogle ScholarPubMed
Esau, C., Davis, S., Murray, S. F., Yu, X. X., Pandey, S. K., Pear, M., Watts, L., Booten, S. L., Graham, M., McKay, R., Subramaniam, A., Propp, S., Lollo, B. A., Freier, S., Bennett, C. F., Bhanot, S. and Monia, B. P. (2006). miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metabolism 3, 8798.CrossRefGoogle ScholarPubMed
Filipowicz, W., Bhattacharyya, S. N. and Sonenberg, N. (2008). Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nature Reviews Genetics 9, 102114.CrossRefGoogle ScholarPubMed
Flynt, A. S. and Lai, E. C. (2008). Biological principles of microRNA-mediated regulation: shared themes amid diversity. Nature Reviews Genetics 9, 831842.CrossRefGoogle ScholarPubMed
Freitas, T. C., Jung, E. and Pearce, E. J. (2007). TGF-beta signaling controls embryo development in the parasitic flatworm Schistosoma mansoni. PLoS Pathogens 3, e52.CrossRefGoogle ScholarPubMed
Fukuda, T., Yamagata, K., Fujiyama, S., Matsumoto, T., Koshida, I., Yoshimura, K., Mihara, M., Naitou, M., Endoh, H., Nakamura, T., Akimoto, C., Yamamoto, Y., Katagiri, T., Foulds, C., Takezawa, S., Kitagawa, H., Takeyama, K., O'Malley, B. W. and Kato, S. (2007). DEAD-box RNA helicase subunits of the Drosha complex are required for processing of rRNA and a subset of microRNAs. Nature Cell Biology 9, 604611.CrossRefGoogle Scholar
Gomes, M. S., Cabral, F. J., Jannotti-Passos, L. K., Carvalho, O., Rodrigues, V., Baba, E. H. and Sa, R. G. (2009). Preliminary analysis of miRNA pathway in Schistosoma mansoni. Parasitology International 58, 6168.CrossRefGoogle ScholarPubMed
Goodchild, J. (2004). Oligonucleotide therapeutics: 25 years agrowing. Current Opinion in Molecular Therapeutics 6, 120128.Google ScholarPubMed
Hao, L., Cai, P., Jiang, N., Wang, H. and Chen, Q. (2010). Identification and characterization of microRNAs and endogenous siRNAs in Schistosoma japonicum. BMC Genomics 11, 55.CrossRefGoogle ScholarPubMed
He, L., He, X., Lim, L. P., de Stanchina, E., Xuan, Z., Liang, Y., Xue, W., Zender, L., Magnus, J., Ridzon, D., Jackson, A. L., Linsley, P. S., Chen, C., Lowe, S. W., Cleary, M. A. and Hannon, G. J. (2007). A microRNA component of the p53 tumour suppressor network. Nature 447, 11301134.CrossRefGoogle ScholarPubMed
Hotez, P. J., Brindley, P. J., Bethony, J. M., King, C. H., Pearce, E. J. and Jacobson, J. (2008). Helminth infections: the great neglected tropical diseases. Journal of Clinical Investigation 118, 13111321.CrossRefGoogle ScholarPubMed
Hu, M., Xia, M., Chen, X., Lin, Z., Xu, Y., Ma, Y. and Su, L. (2010). MicroRNA-141 regulates Smad interacting protein 1 (SIP1) and inhibits migration and invasion of colorectal cancer cells. Digestive Diseases and Sciences 55, 23652372.CrossRefGoogle ScholarPubMed
Hu, W., Yan, Q., Shen, D. K., Liu, F., Zhu, Z. D., Song, H. D., Xu, X. R., Wang, Z. J., Rong, Y. P., Zeng, L. C., Wu, J., Zhang, X., Wang, J. J., Xu, X. N., Wang, S. Y., Fu, G., Zhang, X. L., Wang, Z. Q., Brindley, P. J., McManus, D. P., Xue, C. L., Feng, Z., Chen, Z. and Han, Z. G. (2003). Evolutionary and biomedical implications of a Schistosoma japonicum complementary DNA resource. Nature Genetics 35, 139147.CrossRefGoogle ScholarPubMed
Huang, J., Hao, P., Chen, H., Hu, W., Yan, Q., Liu, F. and Han, Z. G. (2009). Genome-wide identification of Schistosoma japonicum microRNAs using a deep-sequencing approach. PLoS One 4, e8206.CrossRefGoogle ScholarPubMed
Humphreys, D. T., Westman, B. J., Martin, D. I. and Preiss, T. (2005). MicroRNAs control translation initiation by inhibiting eukaryotic initiation factor 4E/cap and poly(A) tail function. Proceedings of the National Academy of Sciences, USA 102, 1696116966.CrossRefGoogle ScholarPubMed
Hutvagner, G., Simard, M. J., Mello, C. C. and Zamore, P. D. (2004). Sequence-specific inhibition of small RNA function. PLoS Biology 2, E98.CrossRefGoogle ScholarPubMed
Hynes, N. E. and MacDonald, G. (2009). ErbB receptors and signaling pathways in cancer. Current Opinion in Cell Biology 21, 177184.CrossRefGoogle ScholarPubMed
Hyun, S., Lee, J. H., Jin, H., Nam, J., Namkoong, B., Lee, G., Chung, J. and Kim, V. N. (2009). Conserved MicroRNA miR-8/miR-200 and its target USH/FOG2 control growth by regulating PI3 K. Cell 139, 10961108.CrossRefGoogle Scholar
Ibanez-Ventoso, C., Vora, M. and Driscoll, M. (2008). Sequence relationships among C. elegans, D. melanogaster and human microRNAs highlight the extensive conservation of microRNAs in biology. PLoS One 3, e2818.CrossRefGoogle ScholarPubMed
Ismail, M., Botros, S., Metwally, A., William, S., Farghally, A., Tao, L. F., Day, T. A. and Bennett, J. L. (1999). Resistance to praziquantel: direct evidence from Schistosoma mansoni isolated from Egyptian villagers. American Journal of Tropical Medicine and Hygiene 60, 932935.CrossRefGoogle ScholarPubMed
Jiang, L., Liu, X., Chen, Z., Jin, Y., Heidbreder, C. E., Kolokythas, A., Wang, A., Dai, Y. and Zhou, X. (2010). MicroRNA-7 targets IGF1R (insulin-like growth factor 1 receptor) in tongue squamous cell carcinoma cells. Biochemical Journal 432, 199205.CrossRefGoogle ScholarPubMed
Johnson, S. M., Grosshans, H., Shingara, J., Byrom, M., Jarvis, R., Cheng, A., Labourier, E., Reinert, K. L., Brown, D. and Slack, F. J. (2005). RAS is regulated by the let-7 microRNA family. Cell 120, 635647.CrossRefGoogle ScholarPubMed
Junn, E., Lee, K. W., Jeong, B. S., Chan, T. W., Im, J. Y. and Mouradian, M. M. (2009). Repression of alpha-synuclein expression and toxicity by microRNA-7. Proceedings of the National Academy of Sciences, USA 106, 1305213057.CrossRefGoogle ScholarPubMed
Kapsimali, M., Kloosterman, W. P., de Bruijn, E., Rosa, F., Plasterk, R. H. and Wilson, S. W. (2007). MicroRNAs show a wide diversity of expression profiles in the developing and mature central nervous system. Genome Biology 8, R173.CrossRefGoogle ScholarPubMed
Karres, J. S., Hilgers, V., Carrera, I., Treisman, J. and Cohen, S. M. (2007). The conserved microRNA miR-8 tunes atrophin levels to prevent neurodegeneration in Drosophila. Cell 131, 136145.CrossRefGoogle ScholarPubMed
Kefas, B., Godlewski, J., Comeau, L., Li, Y., Abounader, R., Hawkinson, M., Lee, J., Fine, H., Chiocca, E. A., Lawler, S. and Purow, B. (2008). microRNA-7 inhibits the epidermal growth factor receptor and the Akt pathway and is down-regulated in glioblastoma. Cancer Research 68, 35663572.CrossRefGoogle ScholarPubMed
Kennell, J. A., Gerin, I., MacDougald, O. A. and Cadigan, K. M. (2008). The microRNA miR-8 is a conserved negative regulator of Wnt signaling. Proceedings of the National Academy of Sciences, USA 105, 1541715422.CrossRefGoogle ScholarPubMed
Khayath, N., Vicogne, J., Ahier, A., BenYounes, A., Konrad, C., Trolet, J., Viscogliosi, E., Brehm, K. and Dissous, C. (2007). Diversification of the insulin receptor family in the helminth parasite Schistosoma mansoni. FEBS Journal 274, 659676.CrossRefGoogle ScholarPubMed
Kim, D. H., Saetrom, P., Snove, O. Jr. and Rossi, J. J. (2008). MicroRNA-directed transcriptional gene silencing in mammalian cells. Proceedings of the National Academy of Sciences, USA 105, 1623016235.CrossRefGoogle ScholarPubMed
Kim, V. N. (2005). MicroRNA biogenesis: coordinated cropping and dicing. Nature Reviews Molecular Cell Biology 6, 376385.CrossRefGoogle ScholarPubMed
Kloosterman, W. P., Lagendijk, A. K., Ketting, R. F., Moulton, J. D. and Plasterk, R. H. (2007). Targeted inhibition of miRNA maturation with morpholinos reveals a role for miR-375 in pancreatic islet development. PLoS Biology 5, e203.CrossRefGoogle ScholarPubMed
Krichevsky, A. M., Sonntag, K. C., Isacson, O. and Kosik, K. S. (2006). Specific microRNAs modulate embryonic stem cell-derived neurogenesis. Stem Cells 24, 857864.CrossRefGoogle ScholarPubMed
Krutzfeldt, J., Rajewsky, N., Braich, R., Rajeev, K. G., Tuschl, T., Manoharan, M. and Stoffel, M. (2005). Silencing of microRNAs in vivo with ‘antagomirs’. Nature 438, 685689.CrossRefGoogle ScholarPubMed
Kwon, C., Han, Z., Olson, E. N. and Srivastava, D. (2005). MicroRNA1 influences cardiac differentiation in Drosophila and regulates Notch signaling. Proceedings of the National Academy of Sciences, USA 102, 1898618991.CrossRefGoogle ScholarPubMed
La Rocca, G., Badin, M., Shi, B., Xu, S. Q., Deangelis, T., Sepp-Lorenzinoi, L. and Baserga, R. (2009). Mechanism of growth inhibition by MicroRNA 145: the role of the IGF-I receptor signaling pathway. Journal of Cellular Physiology 220, 485491.CrossRefGoogle ScholarPubMed
Lagos-Quintana, M., Rauhut, R., Yalcin, A., Meyer, J., Lendeckel, W. and Tuschl, T. (2002). Identification of tissue-specific microRNAs from mouse. Current Biology 12, 735739.CrossRefGoogle ScholarPubMed
Lee, R. C. and Ambros, V. (2001). An extensive class of small RNAs in Caenorhabditis elegans. Science 294, 862864.CrossRefGoogle ScholarPubMed
Lee, R. C., Feinbaum, R. L. and Ambros, V. (1993). The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75, 843854.CrossRefGoogle Scholar
Lee, Y., Ahn, C., Han, J., Choi, H., Kim, J., Yim, J., Lee, J., Provost, P., Radmark, O., Kim, S. and Kim, V. N. (2003). The nuclear RNase III Drosha initiates microRNA processing. Nature 425, 415419.CrossRefGoogle ScholarPubMed
Lee, Y., Kim, M., Han, J., Yeom, K. H., Lee, S., Baek, S. H. and Kim, V. N. (2004). MicroRNA genes are transcribed by RNA polymerase II. The EMBO Journal 23, 40514060.CrossRefGoogle ScholarPubMed
Lewis, B. P., Burge, C. B. and Bartel, D. P. (2005). Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120, 1520.CrossRefGoogle ScholarPubMed
Li, K. K., Pang, J. C., Ching, A. K., Wong, C. K., Kong, X., Wang, Y., Zhou, L., Chen, Z. and Ng, H. K. (2009). miR-124 is frequently down-regulated in medulloblastoma and is a negative regulator of SLC16A1. Human Pathology 40, 12341243.CrossRefGoogle ScholarPubMed
Li, Q., Song, X. W., Zou, J., Wang, G. K., Kremneva, E., Li, X. Q., Zhu, N., Sun, T., Lappalainen, P., Yuan, W. J., Qin, Y. W. and Jing, Q. (2010). Attenuation of microRNA-1 derepresses the cytoskeleton regulatory protein twinfilin-1 to provoke cardiac hypertrophy. Journal of Cell Science 123, 24442452.CrossRefGoogle ScholarPubMed
Lim, L. P., Lau, N. C., Garrett-Engele, P., Grimson, A., Schelter, J. M., Castle, J., Bartel, D. P., Linsley, P. S. and Johnson, J. M. (2005). Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 433, 769773.CrossRefGoogle ScholarPubMed
Liu, K., Liu, Y., Mo, W., Qiu, R., Wang, X., Wu, J. Y. and He, R. (2010). MiR-124 regulates early neurogenesis in the optic vesicle and forebrain, targeting NeuroD1. Nucleic Acids Research 39, 28692879.CrossRefGoogle ScholarPubMed
Loverde, P. T., Osman, A. and Hinck, A. (2007). Schistosoma mansoni: TGF-beta signaling pathways. Experimental Parasitology 117, 304317.CrossRefGoogle ScholarPubMed
Makeyev, E. V., Zhang, J., Carrasco, M. A. and Maniatis, T. (2007). The MicroRNA miR-124 promotes neuronal differentiation by triggering brain-specific alternative pre-mRNA splicing. Molecular Cell 27, 435448.CrossRefGoogle ScholarPubMed
Meyers, B. C., Axtell, M. J., Bartel, B., Bartel, D. P., Baulcombe, D., Bowman, J. L., Cao, X., Carrington, J. C., Chen, X., Green, P. J., Griffiths-Jones, S., Jacobsen, S. E., Mallory, A. C., Martienssen, R. A., Poethig, R. S., Qi, Y., Vaucheret, H., Voinnet, O., Watanabe, Y., Weigel, D. and Zhu, J. K. (2008). Criteria for annotation of plant MicroRNAs. The Plant Cell 20, 31863190.CrossRefGoogle ScholarPubMed
Moretti, F., Thermann, R. and Hentze, M. W. (2010). Mechanism of translational regulation by miR-2 from sites in the 5′ untranslated region or the open reading frame. RNA 16, 24932502.CrossRefGoogle ScholarPubMed
Nelson, P. T., Baldwin, D. A., Kloosterman, W. P., Kauppinen, S., Plasterk, R. H. and Mourelatos, Z. (2006). RAKE and LNA-ISH reveal microRNA expression and localization in archival human brain. RNA 12, 187191.CrossRefGoogle ScholarPubMed
Nohata, N., Sone, Y., Hanazawa, T., Fuse, M., Kikkawa, N., Yoshino, H., Chiyomaru, T., Kawakami, K., Enokida, H., Nakagawa, M., Shozu, M., Okamoto, Y. and Seki, N. (2011). miR-1 as a tumor suppressive microRNA targeting TAGLN2 in head and neck squamous cell carcinoma. Oncotarget 2, 2942.CrossRefGoogle ScholarPubMed
Okamura, K., Hagen, J. W., Duan, H., Tyler, D. M. and Lai, E. C. (2007). The mirtron pathway generates microRNA-class regulatory RNAs in Drosophila. Cell 130, 89100.CrossRefGoogle ScholarPubMed
Osman, A., Niles, E. G., Verjovski-Almeida, S. and LoVerde, P. T. (2006). Schistosoma mansoni TGF-beta receptor II: role in host ligand-induced regulation of a schistosome target gene. PLoS Pathogens 2, e54.CrossRefGoogle ScholarPubMed
Pasquinelli, A. E., Reinhart, B. J., Slack, F., Martindale, M. Q., Kuroda, M. I., Maller, B., Hayward, D. C., Ball, E. E., Degnan, B., Muller, P., Spring, J., Srinivasan, A., Fishman, M., Finnerty, J., Corbo, J., Levine, M., Leahy, P., Davidson, E. and Ruvkun, G. (2000). Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature 408, 8689.CrossRefGoogle ScholarPubMed
Pereira, T. C., Pascoal, V. D., Marchesini, R. B., Maia, I. G., Magalhaes, L. A., Zanotti-Magalhaes, E. M. and Lopes-Cendes, I. (2008). Schistosoma mansoni: evaluation of an RNAi-based treatment targeting HGPRTase gene. Experimental Parasitology 118, 619623.CrossRefGoogle ScholarPubMed
Pierson, J., Hostager, B., Fan, R. and Vibhakar, R. (2008). Regulation of cyclin dependent kinase 6 by microRNA 124 in medulloblastoma. Journal of Neuro-Oncology 90, 17.CrossRefGoogle ScholarPubMed
Poy, M. N., Eliasson, L., Krutzfeldt, J., Kuwajima, S., Ma, X., Macdonald, P. E., Pfeffer, S., Tuschl, T., Rajewsky, N., Rorsman, P. and Stoffel, M. (2004). A pancreatic islet-specific microRNA regulates insulin secretion. Nature 432, 226230.CrossRefGoogle ScholarPubMed
Qin, W., Shi, Y., Zhao, B., Yao, C., Jin, L., Ma, J. and Jin, Y. (2010). miR-24 regulates apoptosis by targeting the open reading frame (ORF) region of FAF1 in cancer cells. PLoS One 5, e9429.CrossRefGoogle ScholarPubMed
Reddy, S. D., Ohshiro, K., Rayala, S. K. and Kumar, R. (2008). MicroRNA-7, a homeobox D10 target, inhibits p21-activated kinase 1 and regulates its functions. Cancer Research 68, 81958200.CrossRefGoogle ScholarPubMed
Reinhart, B. J., Slack, F. J., Basson, M., Pasquinelli, A. E., Bettinger, J. C., Rougvie, A. E., Horvitz, H. R. and Ruvkun, G. (2000). The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature 403, 901906.CrossRefGoogle ScholarPubMed
Ruby, J. G., Jan, C. H. and Bartel, D. P. (2007). Intronic microRNA precursors that bypass Drosha processing. Nature 448, 8386.CrossRefGoogle ScholarPubMed
Savioli, L., Stansfield, S., Bundy, D. A., Mitchell, A., Bhatia, R., Engels, D., Montresor, A., Neira, M. and Shein, A. M. (2002). Schistosomiasis and soil-transmitted helminth infections: forging control efforts. Transactions of the Royal Society of Tropical Medicine and Hygiene 96, 577579.CrossRefGoogle ScholarPubMed
Scherr, M., Venturini, L., Battmer, K., Schaller-Schoenitz, M., Schaefer, D., Dallmann, I., Ganser, A. and Eder, M. (2007). Lentivirus-mediated antagomir expression for specific inhibition of miRNA function. Nucleic Acids Research 35, e149.CrossRefGoogle ScholarPubMed
Shoemaker, C. B., Ramachandran, H., Landa, A., dos Reis, M. G. and Stein, L. D. (1992). Alternative splicing of the Schistosoma mansoni gene encoding a homologue of epidermal growth factor receptor. Molecular and Biochemical Parasitology 53, 1732.CrossRefGoogle ScholarPubMed
Simoes, M. C., Lee, J., Djikeng, A., Cerqueira, G. C., Zerlotini, A., da Silva-Pereira, R. A., Dalby, A. R., LoVerde, P., El-Sayed, N. M. and Oliveira, G. (2011). Identification of Schistosoma mansoni microRNAs. BMC Genomics 12, 47.CrossRefGoogle ScholarPubMed
Simon, D. J., Madison, J. M., Conery, A. L., Thompson-Peer, K. L., Soskis, M., Ruvkun, G. B., Kaplan, J. M. and Kim, J. K. (2008). The microRNA miR-1 regulates a MEF-2-dependent retrograde signal at neuromuscular junctions. Cell 133, 903915.CrossRefGoogle ScholarPubMed
Taguchi, A. and White, M. F. (2008). Insulin-like signaling, nutrient homeostasis, and life span. Annual Review of Physiology 70, 191212.CrossRefGoogle ScholarPubMed
Takane, K., Fujishima, K., Watanabe, Y., Sato, A., Saito, N., Tomita, M. and Kanai, A. (2010). Computational prediction and experimental validation of evolutionarily conserved microRNA target genes in bilaterian animals. BMC Genomics 11, 101.CrossRefGoogle ScholarPubMed
Trabucchi, M., Briata, P., Garcia-Mayoral, M., Haase, A. D., Filipowicz, W., Ramos, A., Gherzi, R. and Rosenfeld, M. G. (2009). The RNA-binding protein KSRP promotes the biogenesis of a subset of microRNAs. Nature 459, 10101014.CrossRefGoogle ScholarPubMed
Vallejo, D. M., Caparros, E. and Dominguez, M. (2011). Targeting Notch signalling by the conserved miR-8/200 microRNA family in development and cancer cells. EMBO Journal 30, 756769.CrossRefGoogle ScholarPubMed
Vicogne, J., Cailliau, K., Tulasne, D., Browaeys, E., Yan, Y. T., Fafeur, V., Vilain, J. P., Legrand, D., Trolet, J. and Dissous, C. (2004). Conservation of epidermal growth factor receptor function in the human parasitic helminth Schistosoma mansoni. Journal of Biological Chemistry 279, 3740737414.CrossRefGoogle ScholarPubMed
Visvanathan, J., Lee, S., Lee, B., Lee, J. W. and Lee, S. K. (2007). The microRNA miR-124 antagonizes the anti-neural REST/SCP1 pathway during embryonic CNS development. Genes & Development 21, 744749.CrossRefGoogle ScholarPubMed
Viswanathan, S. R., Daley, G. Q. and Gregory, R. I. (2008). Selective blockade of microRNA processing by Lin28. Science 320, 97100.CrossRefGoogle ScholarPubMed
Wang, Z., Xue, X., Sun, J., Luo, R., Xu, X., Jiang, Y., Zhang, Q. and Pan, W. (2010). An “in-depth” description of the small non-coding RNA population of Schistosoma japonicum schistosomulum. PLoS Neglected Tropical Diseases 4, e596.CrossRefGoogle ScholarPubMed
Webster, R. J., Giles, K. M., Price, K. J., Zhang, P. M., Mattick, J. S. and Leedman, P. J. (2009). Regulation of epidermal growth factor receptor signaling in human cancer cells by microRNA-7. Journal of Biological Chemistry 284, 57315741.CrossRefGoogle ScholarPubMed
Woods, K., Thomson, J. M. and Hammond, S. M. (2007). Direct regulation of an oncogenic micro-RNA cluster by E2F transcription factors. Journal of Biological Chemistry 282, 21302134.CrossRefGoogle ScholarPubMed
Wurdinger, T. and Costa, F. F. (2007). Molecular therapy in the microRNA era. Pharmacogenomics Journal 7, 297304.CrossRefGoogle ScholarPubMed
Xie, C., Huang, H., Sun, X., Guo, Y., Hamblin, M., Ritchie, R. P., Garcia-Barrio, M. T., Zhang, J. and Chen, Y. E. (2011). MicroRNA-1 regulates smooth muscle cell differentiation by repressing Kruppel-like factor 4. Stem Cells and Development 20, 205210.CrossRefGoogle ScholarPubMed
Xu, C., Lu, Y., Pan, Z., Chu, W., Luo, X., Lin, H., Xiao, J., Shan, H., Wang, Z. and Yang, B. (2007). The muscle-specific microRNAs miR-1 and miR-133 produce opposing effects on apoptosis by targeting HSP60, HSP70 and caspase-9 in cardiomyocytes. Journal of Cell Science 120, 30453052.CrossRefGoogle ScholarPubMed
Xue, X., Sun, J., Zhang, Q., Wang, Z., Huang, Y. and Pan, W. (2008). Identification and characterization of novel microRNAs from Schistosoma japonicum. PLoS One 3, e4034.CrossRefGoogle ScholarPubMed
Yan, D., Dong Xda, E., Chen, X., Wang, L., Lu, C., Wang, J., Qu, J. and Tu, L. (2009). MicroRNA-1/206 targets c-Met and inhibits rhabdomyosarcoma development. Journal of Biological Chemistry 284, 2959629604.CrossRefGoogle ScholarPubMed
Yang, B., Lin, H., Xiao, J., Lu, Y., Luo, X., Li, B., Zhang, Y., Xu, C., Bai, Y., Wang, H., Chen, G. and Wang, Z. (2007). The muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nature Medicine 13, 486491.CrossRefGoogle ScholarPubMed
Yang, Y. P., Guo, S. X., Chen, J., Lin, J. J., Liu, Z. P. and Cheng, G. F. (2010). Cloning, expressing and identifying of a full-length cDNA encoding Argonaute from Schistosoma japonicum. Chinese Journal of Zoonoses 26, 830835.Google Scholar
Yao, G., Yin, M., Lian, J., Tian, H., Liu, L., Li, X. and Sun, F. (2010). MicroRNA-224 is involved in transforming growth factor-beta-mediated mouse granulosa cell proliferation and granulosa cell function by targeting Smad4. Molecular Endocrinology 24, 540551.CrossRefGoogle ScholarPubMed
Yoshino, H., Chiyomaru, T., Enokida, H., Kawakami, K., Tatarano, S., Nishiyama, K., Nohata, N., Seki, N. and Nakagawa, M. (2011). The tumour-suppressive function of miR-1 and miR-133a targeting TAGLN2 in bladder cancer. British Journal of Cancer 104, 808818.CrossRefGoogle ScholarPubMed
You, H., Gobert, G. N., Jones, M. K., Zhang, W. and McManus, D. P. (2011). Signalling pathways and the host-parasite relationship: putative targets for control interventions against schistosomiasis: signalling pathways and future anti-schistosome therapies. Bioessays 33, 203214.CrossRefGoogle ScholarPubMed
You, H., Zhang, W., Jones, M. K., Gobert, G. N., Mulvenna, J., Rees, G., Spanevello, M., Blair, D., Duke, M., Brehm, K. and McManus, D. P. (2010). Cloning and characterisation of Schistosoma japonicum insulin receptors. PLoS One 5, e9868.CrossRefGoogle ScholarPubMed
Yuan, B., Dong, R., Shi, D., Zhou, Y., Zhao, Y., Miao, M. and Jiao, B. (2011). Down-regulation of miR-23b may contribute to activation of the TGF-beta1/Smad3 signalling pathway during the termination stage of liver regeneration. FEBS Letters 585, 927934.CrossRefGoogle ScholarPubMed
Zhao, Z. R., Lei, L., Liu, M., Zhu, S. C., Ren, C. P., Wang, X. N. and Shen, J. J. (2008). Schistosoma japonicum: inhibition of Mago nashi gene expression by shRNA-mediated RNA interference. Experimental Parasitology 119, 379384.CrossRefGoogle ScholarPubMed
Zhou, Y., Zheng, H., Chen, Y., Zhang, L., Wang, K., Guo, J., Huang, Z., Zhang, B., Huang, W., Jin, K., Dou, T., Hasegawa, M., Wang, L., Zhang, Y., Zhou, J., Tao, L., Cao, Z., Li, Y., Vinar, T., Brejova, B., Brown, D., Li, M., Miller, D. J., Blair, D., Zhong, Y., Chen, Z., Liu, F., Hu, W., Wang, Z. Q., Zhang, Q. H., Song, H. D., Chen, S., Xu, X., Xu, B., Ju, C., Huang, Y., Brindley, P. J., McManus, D. P., Feng, Z., Han, Z. G., Lu, G., Ren, S., Wang, Y., Gu, W., Kang, H., Chen, J., Chen, X., Chen, S., Wang, L., Yan, J., Wang, B., Lv, X., Jin, L., Wang, B., Pu, S., Zhang, X., Zhang, W., Hu, Q., Zhu, G., Wang, J., Yu, J., Wang, J., Yang, H., Ning, Z., Beriman, M., Wei, C. L., Ruan, Y., Zhao, G., Wang, S., Liu, F., Zhou, Y., Wang, Z. Q., Lu, G., Zheng, H., Brindley, P. J., McManus, D. P., Blair, D., Zhang, Q. H., Zhong, Y., Wang, S., Han, Z. G., Chen, Z., Wang, S., Han, Z. G. and Chen, Z. (2009). The Schistosoma japonicum genome reveals features of host-parasite interplay. Nature 460, 345351.Google Scholar
Figure 0

Table 1. Putative proteins involved in miRNA biogenesis and its regulation in schistosomes

Figure 1

Table 2. Summary of miRNA discovery in schistosomes

Figure 2

Table 3. Evolutionarily conserved miRNAs in S. japonicum

Figure 3

Table 4. Summary of the targets of evolutionarily conserved miRNAs for schistosomes

Figure 4

Table 5. List of some putative evolutionarily conserved miRNA:mRNA target pairs in S. japonicum