DOI QR코드

DOI QR Code

RNAi and miRNA in Viral Infections and Cancers

  • Published : 2013.12.31

Abstract

Since the first report of RNA interference (RNAi) less than a decade ago, this type of molecular intervention has been introduced to repress gene expression in vitro and also for in vivo studies in mammals. Understanding the mechanisms of action of synthetic small interfering RNAs (siRNAs) underlies use as therapeutic agents in the areas of cancer and viral infection. Recent studies have also promoted different theories about cell-specific targeting of siRNAs. Design and delivery strategies for successful treatment of human diseases are becomingmore established and relationships between miRNA and RNAi pathways have been revealed as virus-host cell interactions. Although both are well conserved in plants, invertebrates and mammals, there is also variabilityand a more complete understanding of differences will be needed for optimal application. RNA interference (RNAi) is rapid, cheap and selective in complex biological systems and has created new insight sin fields of cancer research, genetic disorders, virology and drug design. Our knowledge about the role of miRNAs and siRNAs pathways in virus-host cell interactions in virus infected cells is incomplete. There are different viral diseases but few antiviral drugs are available. For example, acyclovir for herpes viruses, alpha-interferon for hepatitis C and B viruses and anti-retroviral for HIV are accessible. Also cancer is obviously an important target for siRNA-based therapies, but the main problem in cancer therapy is targeting metastatic cells which spread from the original tumor. There are also other possible reservations and problems that might delay or even hinder siRNA-based therapies for the treatment of certain conditions; however, this remains the most promising approach for a wide range of diseases. Clearly, more studies must be done to allow efficient delivery and better understanding of unwanted side effects of siRNA-based therapies. In this review miRNA and RNAi biology, experimental design, anti-viral and anti-cancer effects are discussed.

Keywords

References

  1. Afshar RM, Mollaie H R (2012). Detection of HBV resistance to lamivudine in patients with chronic hepatitis B using Zip nucleic acid probes in Kerman, southeast of Iran. Asian Pac J Cancer Prev, 13, 3657-61. https://doi.org/10.7314/APJCP.2012.13.8.3657
  2. Afshar RM, Mollaie HR (2012). Use of ALLGIO probe assays for detection of HBV resistance to adefovir in patients with chronic hepatitis B, Kerman, Iran. Asian Pac J Cancer Prev, 13, 5463-7. https://doi.org/10.7314/APJCP.2012.13.11.5463
  3. Akashi H, Miyagishi M, Taira K (2004). RNAi expression vectors in plant cells. Methods Mol Biol, 252, 533-43.
  4. Auyeung VC, Ulitsky I, McGeary SE, et al (2013). Beyond secondary structure: primary-sequence determinants license pri-miRNA hairpins for processing. Cell, 152, 844-58. https://doi.org/10.1016/j.cell.2013.01.031
  5. Avci CB, Harman E, Dodurga Y, et al (2013). Therapeutic potential of an anti-diabetic drug, metformin: alteration of miRNA expression in prostate cancer cells. Asian Pac J Cancer Prev, 14, 765-8. https://doi.org/10.7314/APJCP.2013.14.2.765
  6. Bagasra O (2005). RNAi as an antiviral therapy. Expert Opin Biol Ther, 5, 1463-74. https://doi.org/10.1517/14712598.5.11.1463
  7. Barik S (2010). siRNA for influenza therapy. Viruses, 2, 1448-57. https://doi.org/10.3390/v2071448
  8. Boutimah F, Eekels JJ, Liu YP, et al (2013). Antiviral strategies combining antiretroviral drugs with RNAi-mediated attack on HIV-1 and cellular co-factors. Antiviral Res, 98, 121-9. https://doi.org/10.1016/j.antiviral.2013.02.011
  9. Branscheid A, Devers EA, May P, et al (2011). Distribution pattern of small RNA and degradome reads provides information on miRNA gene structure and regulation. Plant Signal Behav, 6, 1609-11. https://doi.org/10.4161/psb.6.10.17305
  10. Broekema NM, Imperiale MJ (2013). miRNA regulation of BK polyomavirus replication during early infection. Proc Natl Acad Sci USA, 110, 8200-5. https://doi.org/10.1073/pnas.1301907110
  11. Cantalupo P, Doering A, Sullivan CS, et al (2005). Complete nucleotide sequence of polyomavirus SA12. J Virol, 79, 13094-104. https://doi.org/10.1128/JVI.79.20.13094-13104.2005
  12. Cao XF, Li SQ (2011). Role of miRNA in the diagnosis, prognosis and treatment of esophageal cancer. Zhonghua Zhong Liu Za Zhi, 33, 161-4.
  13. Castaneda CA, Agullo-Ortuno MT, Fresno Vara, et al (2011). Implication of miRNA in the diagnosis and treatment of breast cancer. Expert Rev Anticancer Ther, 11, 1265-75. https://doi.org/10.1586/era.11.40
  14. Cevec M, Plavec J (2010). Solution structure of miRNA:mRNA complex. Methods Mol Biol, 667, 251-65. https://doi.org/10.1007/978-1-60761-811-9_17
  15. Cevec M, Thibaudeau C, Plavec J (2008). Solution structure of a let-7 miRNA:lin-41 mRNA complex from C elegans. Nucleic Acids Res, 36, 2330-7. https://doi.org/10.1093/nar/gkn088
  16. Chatterjee N, Wang WL, Conklin T, et al (2013). Histone deacetylase inhibitors modulate miRNA and mRNA expression, block metaphase and induce apoptosis in inflammatory breast cancer cells. Cancer Biol Ther, 14, 362-9.
  17. Chaulk SG, Thede GL, Kent OA, et al (2011). Role of pri-miRNA tertiary structure in miR-17-92 miRNA biogenesis. RNA Biol, 8, 1105-14. https://doi.org/10.4161/rna.8.6.17410
  18. Chen J, Zhang W (2012). Kinetic analysis of the effects of target structure on siRNA efficiency. J Chem Phys, 137, 225102. https://doi.org/10.1063/1.4769821
  19. Connelly CM, Uprety R, Hemphill J, et al (2012). Spatiotemporal control of microRNA function using light-activated antagomirs. Mol Biosyst, 8, 2987-93. https://doi.org/10.1039/c2mb25175b
  20. Cruz PE, Mueller C, Cossette TL, et al (2007). In vivo posttranscriptional gene silencing of alpha-1 antitrypsin by adeno-associated virus vectors expressing siRNA. Lab Invest, 87, 893-902. https://doi.org/10.1038/labinvest.3700629
  21. Daniel H, Kim J, John J, et al (2007) .Strategies for silencing human disease using RNA interference. Nature Reviews Genetics, 8, 173-84. https://doi.org/10.1038/nrg2006
  22. De Almeida RS, Keita D, Libeau G, et al (2008). Structure and sequence motifs of siRNA linked with in vitro downregulation of morbillivirus gene expression. Antiviral Res, 79, 37-48. https://doi.org/10.1016/j.antiviral.2008.01.159
  23. Dua P, Yoo J, Kim S, Lee DK (2011). Modified siRNA structure with a single nucleotide bulge overcomes conventional siRNA-mediated off-target silencing. Mol Ther, 19, 1676-87. https://doi.org/10.1038/mt.2011.109
  24. Dzianott A, Sztuba-Solinska J, Bujarski JJ (2012). Mutations in the antiviral RNAi defense pathway modify Brome mosaic virus RNA recombinant profiles. Mol Plant Microbe Interact, 25, 97-106. https://doi.org/10.1094/MPMI-05-11-0137
  25. Eichelser C, Flesch-Janys D, Chang-Claude J, et al (2013). Deregulated Serum Concentrations of Circulating Cell-Free MicroRNAs miR-17, miR-34a, miR-155, and miR-373 in Human Breast Cancer Development and Progression. Clin Chem, 14, 367-72.
  26. Ferrajoli A, Shanafelt TD, Ivan C, et al (2013). Prognostic value of miR-155 in individuals with monoclonal B-cell lymphocytosis and patients with B-chronic lymphocytic leukemia. Blood J, 25, 542-50.
  27. Ferrer-Orta C, Agudo R, Domingo E, et al (2009). Structural insights into replication initiation and elongation processes by the FMDV RNA-dependent RNA polymerase. Curr Opin Struct Biol, 19, 752-8. https://doi.org/10.1016/j.sbi.2009.10.016
  28. Geoghegan JC, Gilmore BL, Davidson BL (2012). Gene Silencing Mediated by siRNA-binding Fusion Proteins Is Attenuated by Double-stranded RNA-binding Domain Structure. Mol Ther Nucleic Acids, 1, 53-5. https://doi.org/10.1038/mtna.2012.43
  29. Gitlin L, Stone JK, Andino R (2005). Poliovirus escape from RNA interference: short interfering RNA-target recognition and implications for therapeutic approaches. J Virol, 79, 1027-35. https://doi.org/10.1128/JVI.79.2.1027-1035.2005
  30. Gorbatyuk OS, Li S, Nash K, et al (2010). In vivo RNAimediated alpha-synuclein silencing induces nigrostriatal degeneration. Mol Ther, 18, 1450-7. https://doi.org/10.1038/mt.2010.115
  31. Gu S, Jin L, Huang Y, et al (2012). Slicing-independent RISC activation requires the argonaute PAZ domain. Curr Biol, 22, 1536-42. https://doi.org/10.1016/j.cub.2012.06.040
  32. Han Q, Zhang C, Zhang J, Tian Z (2011). Involvement of activation of PKR in HBx-siRNA-mediated innate immune effects on HBV inhibition. PLoS One, 6, 27931-35. https://doi.org/10.1371/journal.pone.0027931
  33. Haasnoot J, Westerhout E, Berkhout B (2007). RNA interference against viruses: strike and counterstrike. Nat Biotechnol, 25, 1435-43. https://doi.org/10.1038/nbt1369
  34. He XE, Lei JH, Yang X, et al (2006). [Study of effects of HBV X gene and As2O3 on expression and activity of p53 in HepG2 cells with shRNA]. Zhonghua Gan Zang Bing Za Zhi, 14, 757-61.
  35. He Y, Jiang Y, Wang G, et al (2010). Inhibition of HBV-DNA replication and expression by siRNA based on magnetic nanoparticles transfering in HepG2 2.2.15 cells. Zhong Nan Da Xue Xue Bao Yi Xue Ban, 35, 543-8.
  36. He Y, Sun HQ, He XE, et al (2010). Knockdown of HBx by RNAi inhibits proliferation and enhances chemotherapyinduced apoptosis in hepatocellular carcinoma cells. Med Oncol, 27, 1227-33. https://doi.org/10.1007/s12032-009-9363-0
  37. Hill JM, Zhao Y, Clement C, et al (2009). HSV-1 infection of human brain cells induces miRNA-146a and Alzheimer-type inflammatory signaling. Neuroreport, 20, 1500-5. https://doi.org/10.1097/WNR.0b013e3283329c05
  38. Hong CS, Goins WF, Goss JR, et al (2006). Herpes simplex virus RNAi and neprilysin gene transfer vectors reduce accumulation of Alzheimer's disease-related amyloid-beta peptide in vivo. Gene Ther, 13, 1068-79. https://doi.org/10.1038/sj.gt.3302719
  39. Hosono T, Yokomizo K, Hamasaki A, et al (2008). Antiviral activities against herpes simplex virus type 1 by HPH derivatives and their structure-activity relationships. Bioorg Med Chem Lett, 18, 371-4. https://doi.org/10.1016/j.bmcl.2007.10.065
  40. Hui EK, Yap EM, An DS, et al (2004). Inhibition of influenza virus matrix (M1) protein expression and virus replication by U6 promoter-driven and lentivirus-mediated delivery of siRNA. J Gen Virol, 85, 1877-84. https://doi.org/10.1099/vir.0.79906-0
  41. Idrees S, Ashfaq UA, Khaliq S (2013). RNAi: antiviral therapy against dengue virus. Asian Pac J Trop Biomed, 3, 232-6.
  42. Izquierdo M (2005). Short interfering RNAs as a tool for cancer gene therapy. Cancer Gene Therapy, 12, 217-27. https://doi.org/10.1038/sj.cgt.7700791
  43. Jahan S, Khaliq S, Samreen B, et al (2011). Effect of combined siRNA of HCV E2 gene and HCV receptors against HCV. Virol J, 8, 295-300. https://doi.org/10.1186/1743-422X-8-295
  44. Jahan S, Samreen B, Khaliq S, et al (2011). HCV entry receptors as potential targets for siRNA-based inhibition of HCV. Genet Vaccines Ther, 9, 15-8. https://doi.org/10.1186/1479-0556-9-15
  45. Jankovic R, Radulovic S, Brankovic-Magic M (2009). siRNA and miRNA for the treatment of cancer. J Buon, 14, 43-9.
  46. Jay C, Nemunaitis J, Chen P, et al (2007). miRNA profiling for diagnosis and prognosis of human cancer. DNA Cell Biol, 26, 293-300. https://doi.org/10.1089/dna.2006.0554
  47. Kanasty R, Whitehead K, Vegas A, et al (2012). Action and reaction: the biological response to siRNA and its delivery vehicles. Molecular Therapy, 20,513-24. https://doi.org/10.1038/mt.2011.294
  48. Kayhan B, Yager EJ, Lanzer K, et al (2007). A replicationdeficient murine gamma-herpesvirus blocked in late viral gene expression can establish latency and elicit protective cellular immunity. J Immunol, 179, 8392-402. https://doi.org/10.4049/jimmunol.179.12.8392
  49. Keyvani H, Fazlalipour M, Monavari SH, et al (2012). Hepatitis C virus--proteins, diagnosis, treatment and new approaches for vaccine development. Asian Pac J Cancer Prev, 13, 5931-49.
  50. Kim DH, Rossi JJ (2007). Strategies for silencing human disease using RNA interference. Nat Rev Genet, 8, 173-84. https://doi.org/10.1038/nrg2006
  51. Kim SJ, Ha JW, Zhang BT (2013). Constructing higher-order miRNA-mRNA interaction networks in prostate cancer via hypergraph-based learning. BMC Syst Biol, 7, 47-55. https://doi.org/10.1186/1752-0509-7-47
  52. Knoll S, Emmrich S, Putzer BM (2013). The E2F1-miRNA cancer progression network. Adv Exp Med Biol, 774, 135-47. https://doi.org/10.1007/978-94-007-5590-1_8
  53. Konishi M, Wu CH, Kaito M, et al (2006). siRNA-resistance in treated HCV replicon cells is correlated with the development of specific HCV mutations. J Viral Hepat, 13, 756-61. https://doi.org/10.1111/j.1365-2893.2006.00752.x
  54. Konishi M, Wu CH, Wu GY (2003). Inhibition of HBV replication by siRNA in a stable HBV-producing cell line. Hepatology, 38, 842-50. https://doi.org/10.1053/jhep.2003.50416
  55. Kozielski KL, Tzeng SY, Green JJ (2013). Bioengineered nanoparticles for siRNA delivery. Wiley Interdiscip Rev Nanomed Nanobiotechnol, 25, 654-61.
  56. Krutzfeldt J, Kuwajima S, Braich R, et al (2007). Specificity, duplex degradation and subcellular localization of antagomirs. Nucleic Acids Res, 35, 2885-92. https://doi.org/10.1093/nar/gkm024
  57. Krutzfeldt J, Rajewsky N, Braich R, et al (2005). Silencing of microRNAs in vivo with 'antagomirs'. Nature, 438, 685-9. https://doi.org/10.1038/nature04303
  58. Lee HC, Chen CY, Au LC (2011). Systemic comparison of repression activity for miRNA and siRNA associated with different types of target sequences. [Comparative Study]. Biochem Biophys Res Commun, 411, 393-6. https://doi.org/10.1016/j.bbrc.2011.06.159
  59. Leite KR, Morais DR, Reis ST, et al (2013). MicroRNA 100: a context dependent miRNA in prostate cancer. Clinics (Sao Paulo), 68,652-63. https://doi.org/10.6061/clinics/2013(05)12
  60. Leucci E, Onnis A, Cocco M, et al (2010). B-cell differentiation in EBV-positive Burkitt lymphoma is impaired at posttranscriptional level by miRNA-altered expression. Int J Cancer, 126, 1316-26.
  61. Lu X, Yang G, Zhang J, et al (2011). The sense strand pre-cleaved RNA duplex mediates an efficient RNA interference with less off-target and immune response effects. Appl Microbiol Biotechnol, 90, 583-9. https://doi.org/10.1007/s00253-010-3065-6
  62. Lukiw WJ, Cui JG, Yuan L, et al (2010). Acyclovir or Abeta42 peptides attenuate HSV-1-induced miRNA-146a levels in human primary brain cells. Neuroreport, 21, 922-7. https://doi.org/10.1097/WNR.0b013e32833da51a
  63. Luo KQ, Chang DC (2004). The gene-silencing efficiency of siRNA is strongly dependent on the local structure of mRNA at the targeted region. Biochem Biophys Res Commun, 318, 303-10. https://doi.org/10.1016/j.bbrc.2004.04.027
  64. Ma Z, Li J, He F, et al (2005). Cationic lipids enhance siRNAmediated interferon response in mice. Biochem Biophys Res Commun, 330, 755-9. https://doi.org/10.1016/j.bbrc.2005.03.041
  65. Mallory AC, Ely L, Smith TH, et al (2001). HC-Pro suppression of transgene silencing eliminates the small RNAs but not transgene methylation or the mobile signal. Plant Cell, 13, 571-83. https://doi.org/10.1105/tpc.13.3.571
  66. Malterer G, Dolken L, Haas J (2011). The miRNA-targetome of KSHV and EBV in human B-cells. RNA Biol, 8, 30-4. https://doi.org/10.4161/rna.8.1.13745
  67. Matthew L (2004). RNAi for plant functional genomics. Comp Funct Genomics, 5, 240-4. https://doi.org/10.1002/cfg.396
  68. Merkling SH, van Rij RP (2013). Beyond RNAi: antiviral defense strategies in Drosophila and mosquito. J Insect Physiol, 59, 159-70. https://doi.org/10.1016/j.jinsphys.2012.07.004
  69. Mitani Y, Roberts DB, Fatani H, et al (2013). MicroRNA Profiling of Salivary Adenoid Cystic Carcinoma: Association of miR-17-92 Upregulation with Poor Outcome. PLoS One, 8, 66778-85. https://doi.org/10.1371/journal.pone.0066778
  70. Monteys AM, Spengler RM, Wan J, et al (2010). Structure and activity of putative intronic miRNA promoters. RNA, 16, 495-505. https://doi.org/10.1261/rna.1731910
  71. Morris KV, Rossi JJ (2006). Antiviral applications of RNAi. Curr Opin Mol Ther, 8, 115-21.
  72. Mueller S, Gausson V, Vodovar N, et al (2010). RNAi-mediated immunity provides strong protection against the negativestrand RNA vesicular stomatitis virus in Drosophila. Proc Natl Acad Sci USA, 107, 19390-5. https://doi.org/10.1073/pnas.1014378107
  73. Munson DJ, Burch AD (2012). A novel miRNA produced during lytic HSV-1 infection is important for efficient replication in tissue culture. Arch Virol, 157, 1677-88. https://doi.org/10.1007/s00705-012-1345-4
  74. Nishimura Y, Mieda H, Ishii J, et al (2013). Targeting cancer cell-specific RNA interference by siRNA delivery using a complex carrier of affibody-displaying bio-nanocapsules and liposomes. J Nanobiotechnology, 11, 19-25. https://doi.org/10.1186/1477-3155-11-19
  75. Otaki M (2007). Generation of recombinant adenovirus expressing siRNA against the L mRNA of measles virus and subacute sclerosing panencephalitis virus. Microbiol Immunol, 51, 985-91. https://doi.org/10.1111/j.1348-0421.2007.tb03995.x
  76. Overhoff M, Alken M, Far RK, et al (2005). Local RNA target structure influences siRNA efficacy: a systematic global analysis. J Mol Biol, 348, 871-81. https://doi.org/10.1016/j.jmb.2005.03.012
  77. Pandya UM, Sandhu R, Li B (2013). Silencing subtelomeric VSGs by Trypanosoma brucei RAP1 at the insect stage involves chromatin structure changes. Nucleic Acids Res, 25, 365-420.
  78. Paterson EL, Kazenwadel J, Bert AG, et al (2013). Downregulation of the miRNA-200 family at the invasive front of colorectal cancers with degraded basement membrane indicates EMT is involved in cancer progression. Neoplasia, 15, 180-91. https://doi.org/10.1593/neo.121828
  79. Prakash S, Malhotra M, Rengaswamy V (2010). Nonviral siRNA delivery for gene silencing in neurodegenerative diseases. Methods Mol Biol, 623, 211-29. https://doi.org/10.1007/978-1-60761-588-0_14
  80. Qiu J, Cosmopoulos K, Pegtel M, et al (2011). A novel persistence associated EBV miRNA expression profile is disrupted in neoplasia. PLoS Pathog, 7, 2193-8.
  81. Rand TA, Petersen S, Du F, Wang X (2005). Argonaute2 cleaves the anti-guide strand of siRNA during RISC activation. Cell, 123, 621-9. https://doi.org/10.1016/j.cell.2005.10.020
  82. Scherr M, Venturini L, Eder M (2010). Lentiviral vectormediated expression of pre-miRNAs and antagomiRs. Methods Mol Biol, 614, 175-85. https://doi.org/10.1007/978-1-60761-533-0_12
  83. Schott G, Mari-Ordonez A, Himber C, et al (2012). Differential effects of viral silencing suppressors on siRNA and miRNA loading support the existence of two distinct cellular pools of ARGONAUTE1. Research Support, Non-U.S. Gov't. EMBO J, 31, 2553-65. https://doi.org/10.1038/emboj.2012.92
  84. Seo YS, Jung ES, Kim JH, et al (2009). Significance of anti-HCV signal-to-cutoff ratio in predicting hepatitis C viremia. Korean J Intern Med, 24, 302-8. https://doi.org/10.3904/kjim.2009.24.4.302
  85. Shlomai A, Lubelsky Y, Har-Noy O, et al (2009). The “Trojan horse” model-delivery of anti-HBV small interfering RNAs by a recombinant HBV vector. Biochem Biophys Res Commun, 390, 619-23. https://doi.org/10.1016/j.bbrc.2009.10.016
  86. Shlomai A, Shaul Y (2004). RNA interference--small RNAs effectively fight viral hepatitis. Liver Int, 24, 526-31. https://doi.org/10.1111/j.1478-3231.2004.0960.x
  87. Shwetha S, Gouthamchandra K, Chandra M, et al (2013).Circulating miRNA profile in HCV infected serum: novelinsight into pathogenesis. Sci Rep, 3, 1555. https://doi.org/10.1038/srep01555
  88. Sihan W, Wenbo Z, Guangmei Y (2013). Evolvement of microRNAs as therapeutic targets for malignant gliomas. INTECH, 2, 27-35.
  89. Sioud M (2010). Advances in RNA sensing by the immune system: separation of siRNA unwanted effects from RNA interference. Methods Mol Biol, 629, 33-52. https://doi.org/10.1007/978-1-60761-657-3_3
  90. Suzuki T, Orba Y, Okada Y, et al (2010). The human polyoma JC virus agnoprotein acts as a viroporin. PLoS Pathog, 6, 801-21.
  91. Takigawa Y, Nagano-Fujii M, Deng L, et al (2004). Suppression of hepatitis C virus replicon by RNA interference directed against the NS3 and NS5B regions of the viral genome. Microbiol Immunol, 48, 591-8. https://doi.org/10.1111/j.1348-0421.2004.tb03556.x
  92. Tang KF, Xie J, Chen M, et al (2008). Knockdown of damagespecific DNA binding protein 1 (DDB1) enhances the HBxsiRNA-mediated inhibition of HBV replication. Biologicals, 36, 177-83. https://doi.org/10.1016/j.biologicals.2007.11.002
  93. Tchernitsa O, Kasajima A, Schafer R, et al (2010). Systematic evaluation of the miRNA-ome and its downstream effects on mRNA expression identifies gastric cancer progression. J Pathol, 222, 310-9. https://doi.org/10.1002/path.2759
  94. Tian W, Liou HC (2009). RNAi-mediated c-Rel silencing leads to apoptosis of B cell tumor cells and suppresses antigenic immune response in vivo. PLoS One, 4, 5028-35. https://doi.org/10.1371/journal.pone.0005028
  95. Tie Y, Liu B, Fu H, et al (2009). Circulating miRNA and cancer diagnosis. Sci China C Life Sci, 52, 1117-22. https://doi.org/10.1007/s11427-009-0158-5
  96. Trejo-Avila L, Elizondo-Gonzalez R, Trujillo-Murillo Kdel, et al (2007). Antiviral therapy: inhibition of Hepatitis C Virus expression by RNA interference directed against the NS5B region of the viral genome. Ann Hepatol, 6, 174-80.
  97. Truong NP, Gu W, Prasadam IJ, et al (2013). An influenza virusinspired polymer system for the timed release of siRNA. Nat Commun, 4, 1902. https://doi.org/10.1038/ncomms2905
  98. Tsutsumi A, Kawamata T, Izumi N, et al (2011). Recognition of the pre-miRNA structure by Drosophila Dicer-1. Nat Struct Mol Biol, 18, 1153-8. https://doi.org/10.1038/nsmb.2125
  99. Umbach JL, Cullen BR (2009). The role of RNAi and microRNAs in animal virus replication and antiviral immunity. Genes Dev, 23, 1151-64. https://doi.org/10.1101/gad.1793309
  100. Van Mierlo JT, Van Cleef KW, Van Rij RP (2011). Defense and counterdefense in the RNAi-based antiviral immune system in insects. Methods Mol Biol, 721, 3-22. https://doi.org/10.1007/978-1-61779-037-9_1
  101. Vlachakis D, Tsiliki G, Pavlopoulou A, et al (2013). Antiviral Stratagems Against HIV-1 Using RNA Interference (RNAi) Technology. Evol Bioinform Online, 9, 203-13.
  102. Wang K, Chen X, Yan F, et al (2013). 5'-Triphosphate-siRNA Against Survivin Gene Induces Interferon Production and Inhibits Proliferation of Lung Cancer Cells In Vitro. J Immunother, 36, 294-304. https://doi.org/10.1097/CJI.0b013e318294183b
  103. Wang Y, Kato N, Jazag A, et al (2006). Hepatitis C virus core protein is a potent inhibitor of RNA silencing-based antiviral response. Gastroenterology, 130, 883-92. https://doi.org/10.1053/j.gastro.2005.12.028
  104. Weiwei M, Zhenhua X, Feng L, et al (2009). A significant increase of RNAi efficiency in human cells by the CMV enhancer with a tRNAlys promoter. J Biomed Biotechnol, 20, 514-20.
  105. Wilson JA, Richardson CD (2006). Future promise of siRNA and other nucleic acid based therapeutics for the treatment of chronic HCV. Infect Disord Drug Targets, 6, 43-56. https://doi.org/10.2174/187152606776056689
  106. Wu CJ, Huang HW, Liu CY, et al (2005). Inhibition of SARSCoV replication by siRNA. Antiviral Res, 65, 45-8. https://doi.org/10.1016/j.antiviral.2004.09.005
  107. Wu J, Nandamuri KM (2004). Inhibition of hepatitis viral replication by siRNA. Expert Opin Biol Ther, 4, 1649-59. https://doi.org/10.1517/14712598.4.10.1649
  108. Wu Y, Huang AL, Tang N, et al (2005). Specific anti-viral effects of RNA interference on replication and expression of hepatitis B virus in mice. Chin Med J (Engl), 118, 1351-6.
  109. Wu YH, Hu TF, Chen YC, et al (2011). The manipulation of miRNA-gene regulatory networks by KSHV induces endothelial cell motility. Blood, 118, 2896-905. https://doi.org/10.1182/blood-2011-01-330589
  110. Xu W, Wang LW, Shi JZ, et al (2009). Effects of RNA interference targeting transforming growth factor-beta 1 on immune hepatic fibrosis induced by Concanavalin A in mice. Hepatobiliary Pancreat Dis Int, 8, 300-8.
  111. Yang SJ, Wang CL, Tang EJ (2010). [Inhibition of HBsAg and HBeAg expression by shRNA expressing vectors targeting HBV s and e gene]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi, 26, 1198-9.
  112. Ye J, Wu X, Wu D, et al (2013). miRNA-27b targets vascular endothelial growth factor C to inhibit tumor progression and angiogenesis in colorectal cancer. PLoS One, 8, 60687-98. https://doi.org/10.1371/journal.pone.0060687
  113. Ye X, Huang N, Liu Y, et al (2011). Structure of C3PO and mechanism of human RISC activation. Nat Struct Mol Biol, 18, 650-7. https://doi.org/10.1038/nsmb.2032
  114. Yokota T, Iijima S, Kubodera T, et al (2007). Efficient regulation of viral replication by siRNA in a non-human primate surrogate model for hepatitis C. Biochem Biophys Res Commun, 361, 294-300. https://doi.org/10.1016/j.bbrc.2007.06.182
  115. Yuan J, Yu M, Lin QW, et al (2010). Neutralization of IL-17 inhibits the production of anti-ANT autoantibodies in CVB3- induced acute viral myocarditis. Int Immunopharmacol, 10, 272-6. https://doi.org/10.1016/j.intimp.2009.11.010
  116. Zhao S, Yao D, Chen J, et al (2013). Circulating miRNA-20a and miRNA-203 for Screening Lymph Node Metastasis in Early Stage Cervical Cancer. Genet Test Mol Biomarkers, 25, 365-70.
  117. Zhou J, Rossi JJ (2011). Progress in RNAi-based antiviral therapeutics. Methods Mol Biol, 721, 67-75. https://doi.org/10.1007/978-1-61779-037-9_4
  118. Zhou J, Rossi JJ (2012). Therapeutic potential of aptamer-siRNA conjugates for treatment of HIV-1. Bio Drugs, 26, 393-400.
  119. Zhu Q, Feng C, Liao W, et al (2013). Target delivery of MYCN siRNA by folate-nanoliposomes delivery system in a metastatic neuroblastoma model. Cancer Cell Int, 13, 65-70. https://doi.org/10.1186/1475-2867-13-65
  120. Zhu S, Jiang Q, Wang G, et al (2011). Chromatin structure characteristics of pre-miRNA genomic sequences. BMC Genomics, 12, 329-35. https://doi.org/10.1186/1471-2164-12-329

Cited by

  1. How to approach and treat viral infections in ICU patients vol.14, pp.1, 2014, https://doi.org/10.1186/1471-2334-14-321
  2. Effects of miR-152 on Cell Growth Inhibition, Motility Suppression and Apoptosis Induction in Hepatocellular Carcinoma Cells vol.15, pp.12, 2014, https://doi.org/10.7314/APJCP.2014.15.12.4969
  3. Coaxing Cancer Pro-Apoptoticity: An Approach Blending Therapeutic miRNAs and Dietary Phytochemicals vol.15, pp.13, 2014, https://doi.org/10.7314/APJCP.2014.15.13.5499
  4. TRAIL Based Therapy: Overview of Mesenchymal Stem Cell Based Delivery and miRNA Controlled Expression of TRAIL vol.15, pp.16, 2014, https://doi.org/10.7314/APJCP.2014.15.16.6495
  5. MicroRNAs in Colorectal Cancer: from Diagnosis to Targeted Therapy vol.15, pp.17, 2014, https://doi.org/10.7314/APJCP.2014.15.17.6989
  6. MicroRNAs: Biogenesis, Roles for Carcinogenesis and as Potential Biomarkers for Cancer Diagnosis and Prognosis vol.15, pp.18, 2014, https://doi.org/10.7314/APJCP.2014.15.18.7489
  7. Mechanisms of miRNA-Mediated Gene Regulation from Common Downregulation to mRNA-Specific Upregulation vol.2014, pp.2314-4378, 2014, https://doi.org/10.1155/2014/970607
  8. Inhibition of c-FLIP by RNAi Enhances Sensitivity of the Human Osteogenic Sarcoma Cell Line U2OS to TRAILInduced Apoptosis vol.16, pp.6, 2015, https://doi.org/10.7314/APJCP.2015.16.6.2251
  9. Identification and bioinformatic analysis of dysregulated microRNAs in human oligodendroglial cells infected with borna disease virus vol.14, pp.5, 2016, https://doi.org/10.3892/mmr.2016.5842
  10. Engineered Hydrogels for Local and Sustained Delivery of RNA-Interference Therapies vol.6, pp.1, 2016, https://doi.org/10.1002/adhm.201601041
  11. Efficacy Analysis of Combinatorial siRNAs against HIV Derived from One Double Hairpin RNA Precursor vol.8, pp.1664-302X, 2017, https://doi.org/10.3389/fmicb.2017.01651
  12. Combined Elevation of microRNA-196a and microRNA-196b in Sera Predicts Unfavorable Prognosis in Patients with Osteosarcomas vol.15, pp.4, 2014, https://doi.org/10.3390/ijms15046544
  13. Impact of size, secondary structure, and counterions on the binding of small ribonucleic acids to layered double hydroxide nanoparticles vol.10, pp.4, 2015, https://doi.org/10.1116/1.4936393
  14. Functional Regulation of an Autographa californica Nucleopolyhedrovirus-Encoded MicroRNA, AcMNPV-miR-1, in Baculovirus Replication vol.90, pp.14, 2016, https://doi.org/10.1128/JVI.00165-16
  15. Dengue virus causes changes of MicroRNA-genes regulatory network revealing potential targets for antiviral drugs vol.12, pp.1, 2018, https://doi.org/10.1186/s12918-017-0518-x
  16. Effects of human cyclooxygenase-2 gene silencing on synovial cells of rheumatoid arthritis mediated by lentivirus pp.2169-141X, 2018, https://doi.org/10.1080/21691401.2018.1491479