Inhibition of HIV-1 replication by RNA with a microRNA-like function

  • Authors:
    • Keijiro Kato
    • Takashi Senoki
    • Hiroshi Takaku
  • View Affiliations

  • Published online on: November 1, 2012     https://doi.org/10.3892/ijmm.2012.1170
  • Pages: 252-258
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Human immunodeficiency virus type 1 (HIV-1) replication is suppressed by a small guide RNA (sgRNA) that targets the packaging signal of HIV-1 RNA. We unintentionally produced a plasmid with the reverse sequence of the sgRNA and its terminator (pR-Ψ-sgRNA-ter). Both sgRNA and R-Ψ-sgRNA suppress HIV-1, but the mechanism by which R-Ψ-sgRNA suppresses HIV is not clear. To evaluate whether the suppressive effect is caused by an RNA interference or microRNA (miRNA)-like mechanism, R-Ψ-sgRNA was synthesized in vitro and treated with the Dicer enzyme, an important enzyme for RNA interference and miRNA. The RNA was cleaved into fragments of approximately 24 nucleotides (nt). We analyzed the sequence of the RNA fragments and predicted the RNA secondary structure of R-Ψ-sgRNA to determine the region recognized by the Dicer enzyme. The lengths of the R-Ψ-sgRNA fragments ranged from 48 to 140 nt, and were predicted to form double strands, including mismatches, in this region. An HIV-1 p24 assay indicated that the R-Ψ-sgRNA fragments suppressed HIV-1 replication. These findings suggest that R-Ψ-sgRNA acts as a miRNA to inhibit HIV-1.

Introduction

RNA interference (RNAi) refers to the sequence-specific degradation of RNA that follows the cellular introduction of homologous, short interfering RNA (siRNA) (14). RNAi has emerged as a powerful tool for probing the function of genes of a known sequence both in vitro and in vivo. Advances in vector design permit the effective expression of siRNA in human cells by transferring short hairpin RNA (shRNA) expression cassettes. Recent studies reported the ability of RNAi to decrease replication of human immunodeficiency virus type 1 (HIV-1) in lymphocytes using siRNA targeting viral (such as tat, gag, rev, env, nef) (511) and host (such as CCR5, CD4) (1214) proteins. Thus, RNAi might potentially be used as a form of genetic therapy for HIV-1 and associated infections. Another type of short RNA, microRNA (miRNA), has also been reported (1517). miRNAs are transcribed from the genome as the primary miRNA and digested by Drosha RNase III enzyme to produce the precursor-miRNA in the nucleus. The precursor miRNA is then transported to the cytoplasm and digested by Dicer to become mature miRNA. The miRNA is incorporated into RISC and binds to the target RNA, similar to siRNA (1822). Since the miRNA has mismatches with the target sequence, translation of the target RNA is suppressed. A large number of miRNAs was recently identified in humans. It is estimated that tens of thousands of miRNAs are coded in the genome, and that these miRNAs control protein expression (18,23). miRNAs are important for maintaining cell functions. New short-strand RNAs have also been found (2433). For example, a piwi-interacting RNA binding a piwi subfamily protein is thought to be involved in silencing retrotransposons in fetal male germ cells via DNA methylation of their regulatory regions (34), whereas endogenous siRNAs from naturally occurring double-stranded RNAs are thought to regulate protein-coding transcripts and retrotransposons in mammalian oocytes (35). Most small RNAs are coded in the non-coding region of the genome, and the more complex the organism, the larger the non-coding region. Further investigation of the functions of the small RNAs is essential for a better understanding of biological systems.

HIV-1 is the most common pathogenic AIDS virus. HIV-1 infects the immune cells by integrating the viral genome into the host cell genome. Viral proteins are expressed by the integrated genome through the host cell genetic expression mechanisms. Viral particles that bud on the cell membrane are then released from the cells. A packaging signal (Ψ) on HIV-1 mRNA is required for incorporation of the HIV-1 RNA genome into a viral particle when the HIV-1 virus forms on a host cell membrane.

To suppress HIV-1 replication, we constructed a small-guide RNA (sgRNA) expression vector that targets the SL3 of Ψ and evaluated its effect on HIV-1 replication (3638). The sgRNA is approximately 30-nucleotides (nt) long and forms a tRNA-like structure with the target RNA in the nucleus, where it cleaves the target RNA by activating the tRNA processing enzyme tRNAZL (3842).

A plasmid vector with the reverse sequence of the sgRNA and terminator was unintentionally constructed. This reverse-sequence sgRNA expression vector, pR-Ψ-sgRNA-ter, also inhibited HIV-1 replication (Fig. 1) (43). The R-Ψ-sgRNA expressed by the pR-Ψ-sgRNA-ter vector is 200 nt long, and, to suppress HIV-1 replication, must contain the reverse sequence of the sgRNA (sgRNA-R), terminator (ter-R), and also the plasmid sequence. The HIV-1 suppressing mechanism of R-Ψ-sgRNA, however, remains unclear. Thus, to elucidate the mechanism of HIV-1 suppression by R-Ψ-sgRNA, we evaluated the potential involvement of RNAi or miRNA.

Materials and methods

Cell culture and transfections

HeLa CD4+ cells were grown in RPMI-1640 medium (Sigma, St. Louis, MO, USA) supplemented with 10% (v/v) heat-inactivated fetal bovine serum (FBS), 50 U/ml penicillin, and 50 U/ml streptomycin at 37°C in a 5% CO2 atmosphere. Transfection was carried out using the FuGENE™ 6 reagent (Roche Diagnostics, Indianapolis, IN, USA) or DMRIE-C (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s protocol.

Plasmid construction

To construct pR-Ψ-sgRNA-ter, an oligonucleotide consisting of sgRNA and terminator sequences was ligated into the KpnI sites of pL6 vector (38,44) of pSV2neo (L6) (38) with the tRNAiMet promoter (45). One side of the oligonucleotide was designed not to be digested with KpnI when the oligonucleotide was ligated to KpnI sites of pL6 vector. pR-Gag-sgRNA-ter was constructed in the same way as pR-Ψ-sgRNA-ter. A PCR product to construct pR-Ψ-sgRNA-ter-1 was amplified with specific primers (forward, 5′-GGAATTCCAATTCGC AACCTGTGGTAGC-3′ and reverse, 5′-GAAGATCTTCC CCCAAAAAAATAAAGCATTTTTTTCACT-3′) to pR-Ψ-sgRNA-ter. The forward primer had a KpnI restriction site. The reverse primer had a BglII restriction site. The PCR product was ligated to the KpnI and BamHI sites of the pL6 vector. pL6-ter served as a negative control.

Luciferase assay

The pNL4-3lucΔenv (pNL-luc) was an HIV-1 NL4-3-based vector containing a firefly luciferase gene as a reporter (46). The nef gene along with some of the envelope gene sequences of the HIV-1 NL4-3 genome were deleted, and the firefly luciferase gene was inserted. pNL-luc (0.1 μg) and either pL6 plasmid pR-Ψ-sgRNA-ter, pR-Gag-sgRNA-ter, pR-Ψ-sgRNA-ter-1, pter-R+sgRNA-R+ter, psgRNAR, pter-R or pL6-ter serving as a negative control (0.1 μg) were co-transfected with the transfection reagent, FuGENE 6, into HeLa CD4+ cells. A PicaGene kit (Toyo Ink Co., Ltd., Tokyo, Japan) and a Luciferase Assay System (Promega, Madison, WI, USA) were used for the luciferase assay. Two days after transfection, the transfected culture cells were treated with cell lysis buffer. Cell lysate was recovered into 1.5-ml tubes to remove the unused components by centrifugation. Lysate was reacted with luminous substrate and the level of fluorescence was measured on a luminometer (Lumat LB 9507; Berthold Technologies, Bad Wildbad, Germany). The amount of firefly luciferase activity was normalized with reference to the protein concentration of the cell lysate. The protein assay was quantitated using the BCA Protein Assay Reagent kit (Thermo Scientific Inc., Rockford, IL, USA), which is based on bicinchoninic acid (BCA).

RNA synthesis and digestion

The DNA template used to synthesize R-Ψ-sgRNA was amplified with specific primers (forward, 5′-GACTCGTAATACGACTCACTATAGGCAG AACAGCAGAGTGGCG-3′ and reverse, 5′-AATAAAGCAT TTTTTTCACTGCATTCTAGT-3′) to the pR-Ψ-sgRNA-ter by PCR. The forward primer had a T7 promoter sequence to transcribe the R-Ψ-sgRNA. The amplified DNA template was incubated with T7 RNA polymerase (Invitrogen) and nucleotide triphosphates for 3 h at 37°C and the synthesized R-Ψ-sgRNA was treated with DNase I (Takara Bio Co., Ltd., Shiga, Japan) to remove the DNA template. R-Ψ-sgRNA was confirmed by 2.0% denatured formaldehyde agarose gel electrophoresis. R-Ψ-sgRNA was incubated with recombinant human Dicer enzyme of BLOCK-iT™ Dicer RNAi kits (Invitrogen) and purified according to the manufacturer’s protocol. Digested RNA fragments were confirmed by 15% polyacrylamide gel electrophoresis.

Northern blot analysis

Small RNA (<200 nt) was extracted from pR-Ψ-sgRNA-ter and siRNA-Dicer (0, 10, 20, 30 nM) co-transfected HeLa CD4+ cells using a mirVana™ miRNA Isolation kit, according to the manufacturer’s instructions (Ambion, Foster City, CA, USA). siRNA corresponding to Dicer [5′-UGCUUGAAGCAGCUCUGGA(dTdT)-3′] was purchased from Invitrogen. Small RNA 5 μg samples were loaded onto a 15% (w/v) polyacrylamide/7 M urea gel. After transfer to a Hybond-N™ nylon membrane (GE Healthcare Bio-Sciences Corp., Piscataway, NJ, USA), synthetic LNA/DNA oligonucleotides (5′-biotin-TGGATCCCCGGGTACGTCTCC-3′) complementary to the antisense strand of the R-Ψ-sgRNA region (54–75 nt). The membranes were prehybridized for 1 h in North2South hybridization buffer (Pierce) at 55°C and hybridized overnight to the 5′-biotin labeled LNA/DNA probe (30 ng/ml of hybridization buffer). Four posthybridization washes were carried out at 20 min each at 65°C with 2× SSC, 1× SSC is 0.15 M NaCl plus 0.015 M sodium citrate-0.1% sodium dodecyl sulfate (SDS). Detection of LNA/DNA/RNA hybrids was carried out using the North2South chemiluminescent detection system (Thermo Scientific Inc.)

RNA sequence analysis

A poly(A) tail was added to the 3′ terminal ends of the R-Ψ-sgRNA fragments with poly(A) polymerase (Ambion, Austin TX, USA) and subsequently ligated to a 5′ adapter (5′-ACGGAATTCCTCACTaaa-3′; uppercase letters represent DNA, the lowercase letters represent RNA) to the 5′ terminals with T4 RNA ligase (New England Biolabs, Ipswich, MA, USA). Reverse transcription was performed with the poly(A)-specific reverse transcription (RT) primer consisting of a continuous T sequence and an additional sequence to convert RNA to DNA. The RT primer was as follows: 5′-CTGATCTAGAGGT ACCGGATCCTTTTTTTTTTTTTTTTTTTT-3′. ReverTra-α (Toyobo Co., Ltd., Osaka, Japan) was used for this reaction. The additional sequence of the RT primer was used to adjust the annealing temperature of the following PCR. The reverse transcripts were amplified with PCR primers specific to the 5′ adapter sequence (5′-CAGCCAACGGAATTCCTCACTAAA-3′) and the RT primer by PCR. The PCR products were inserted into the cloning vector, pGEM-Easy vector (Promega), for sequence analysis. Sequence analysis was performed with an ABI PRISM 310 Genetic Analyzer (PE Applied Biosystems, Tokyo, Japan) according to the manufacturer’s protocol.

HIV-1 p24 assay

For the HIV-1 p24 assay, HeLa CD4+ cells were plated onto 12-well plates (Iwaki Glass, Tokyo, Japan). After 24 h, the medium was replaced with fresh medium and the cells were transfected with small RNA fragments of R-Ψ-sgRNA [(0.5, 0.25 and 0.05 μg) and HIV-1 pNL4-3 (0.05 μg)]. The transfection agents were DMRIE-C (Invitrogen) and FuGENE 6 (Roche Diagnostics). Two days later, HIV-1 gag p24 antigen levels were measured using a chemiluminescent enzyme immunoassay (Lumipulse; Fujirebio, Tokyo, Japan) (47).

Results and Discussion

Inhibition of HIV-1 replication with the reverse sequence sgRNA expression vector

Recently, we demonstrated the inhibition of HIV-1 gene products in cultured cells by inducing HIV-1 mRNA cleavage using a modified 5′-half-tRNAArg (sgRNA) and mammalian tRNA 3′-processing endoribonuclease (tRNase ZL) (38). The sgRNA/target HIV-1RNA complex formed a pre-tRNA-like structure with 5′-half-tRNA and a stable hairpin (3′-half-tRNA) structure resembling the T stem-loop region (Fig. 1A and B). The tRNAmet-sgRNA transcript was expressed at high levels and localized in the nucleus. The greatest inhibitory effect on HIV-1 expression was achieved using sgRNA targeting the HIV-1 packaging (Ψ) signal gene (3638). To construct the sgRNA expression plasmid, a pL6 vector, dpSV/neo with a human methionine tRNA promoter (38,45), was used to insert the sgRNA and terminator sequences. During this process, pR-Ψ-sgRNA-ter, which has the reverse sequence of the sgRNA and the terminator, was unintentionally constructed (Fig. 1C). A transient-expression assay was used to test the ability of pR-Ψ-sgRNA-ter to inhibit HIV-1 expression. HeLa CD4+ cells were co-transfected with the sgRNA plasmids and an HIV-1NL4-3 based-vector containing a luciferase-expression marker (pNL-luc) (Fig. 2A) (46), and then HIV-1 suppression was assessed with a single-cycle infectivity assay. HIV-1NL4-3 based-luciferase activity was recorded using the control plasmid vector pL6-ter with only the tRNAmet promoter and terminator sequences, rather than the sgRNA-expression plasmid. R-Ψ-sgRNA-ter showed good inhibition of HIV-1 expression in cultured cells (Fig. 2C). The contribution of the R-Ψ-sgRNA-ter to the anti-HIV-1 effect was examined by three types of modified R-Ψ-sgRNA-ter plasmids (R-Ψ-sgRNA-ter that did not contain ter-R+sgRNA-R+ter, sgRNA-R or ter-R) as defective pR-Ψ-sgRNA-ters (Fig. 2B). We also constructed a control R-sgRNA, pR-Gag-sgRNA-ter containing the reverse sequence of the sgRNA targeting the HIV-1 gag gene (gag initiation codon site) (Fig. 2B). The luciferase assay indicated that the modified R-Ψ-sgRNA-ter plasmids without the sgRNA-R, ter-R, or plasmid sequence (ter-R+sgRNA-R+ter) and the control sgRNA, R-Gag-sgRNA-ter did not suppress HIV-1 expression (Fig. 2C). These results suggest that the R-Ψ-sgRNA-ter sequence is critical to HIV-1 specific expression.

Length of R-Ψ-sgRNA and its inhibitory effect on HIV-1 replication

The pR-Ψ-sgRNA-ter termination signal sequence was also reversed, and pR-Ψ-sgRNA-ter did not have a terminator. Therefore we could not predict the length of the R-Ψ-sgRNA transcribed from pR-Ψ-sgRNA-ter. To determine the length of the R-Ψ-sgRNA transcribed from pR-Ψ-sgRNA-ter, an RNA sequence analysis was performed. The total RNA of pR-Ψ-sgRNA-ter transfected cells was treated with poly(A) polymerase to add a poly(A) tail to the 3′ terminal end of R-Ψ-sgRNA. A PCR primer specific to the transcribed tRNAmet promoter sequence and RT primer specific to the poly(A) tail were used for RT-PCR. A cloning vector was used to insert the PCR products, and the sequences were analyzed. RNA sequence analysis revealed that the R-Ψ-sgRNA-ter was 200 nt long (Fig. 3A and B). We confirmed that R-Ψ-sgRNA contained sgRNA-R, ter-R, and a plasmid sequence following sgRNA-R. A plasmid vector expressing the R-Ψ-sgRNA-ter-1 (200 nt) sequence was constructed with pL6 vector, and a luciferase assay indicated that the R-Ψ-sgRNA-ter-1 (200 nt) suppressed HIV-1 expression (Fig. 3C). Size-selected RNAs were electrophoresed in polyacrylamide gels and transferred to nylon membranes. We then hybridized the membranes with a synthetic LNA/DNA oligonucleotide (5′-biotin-TGGATCCCCGGGTACGTCTCC-3′) complementary to the antisense strand of the R-Ψ-sgRNA region (54–75 nt) and detected a 24 bp signal (Fig. 3D, lane 2) not seen in the control-ter-R cells (Fig. 3D, lane 1). Furthermore, HeLa CD4+ cells were transfected with pR-Ψ-sgRNA-ter and Dicer-siRNA (334–352 nt, 10–30 nM) and the expected small RNA decreased in a dose-dependent manner (Fig. 3D, lanes 3–5). These results suggest that the R-Ψ-sgRNA-ter-1 (200 nt) sequence is required for HIV-1 suppression.

Sequence analysis of the digested RNA fragments

To search for the double-stranded region recognized by Dicer, an RNA sequence analysis was performed. A poly(A) tail and 5′ adapter were added to the digested RNA fragments and the RNA was converted to DNA by RT-PCR. A cloning vector was used to insert the DNA fragments and the sequences were analyzed. Most of the RNA fragment sequences existed in two regions of the R-Ψ-sgRNA: the region from ter-R to the anterior sequence of ter-R, which was transcribed initially by the tRNA promoter, and the region from sgRNA-R to the plasmid sequence following sgRNA-R (Fig. 4A). In addition, the sgRNA-R sequences obtained from the sequence analysis were not present in the sgRNA-R region of the pR-Gag-sgRNA-ter targeting gag gene. These results, therefore, seemed to be related to the use of defective pR-Ψ-sgRNA-ter and pR-Gag-sgRNA-ter in the assay (Fig. 2).

The RNA secondary structure of R-Ψ-sgRNA was predicted using the Mfold program (48) and the double-stranded region was searched based on the RNA sequence data obtained from the sequence analysis. The double-stranded region was located between 48 to 140 nt of the R-Ψ-sgRNA, and the presence of sequences of short-strand RNA fragments in this region was confirmed (Fig. 4B). Furthermore, miRNAs recognize HIV-1 NL4-3 target mRNAs by their hybridization pattern as predicted using the miRNA target scanning algorithm program (49). The miRNA region was searched based on the 24 nt small RNA shown in Fig. 3D, and the presence of miRNA in the double-stranded region (nucleotides 48–140) of R-Ψ-sgRN was confirmed (data not shown). The double-stranded region had mismatches, indicating that the short-strand RNA fragments include miRNA, but not siRNA.

Digestion of R-Ψ-sgRNA with Dicer

We hypothesized that the secondary structure of the R-Ψ-sgRNA had double-stranded regions that may be recognized by the Dicer RNase III enzyme, which produces the 24 nt long RNA fragments that are important for RNAi and miRNA. To examine whether the HIV-1 suppression mechanism of R-Ψ-sgRNA involved RNAi or miRNA, R-Ψ-sgRNA was synthesized in vitro and digested with Dicer (50). The size of the digested RNA fragments was confirmed with 15% polyacrylamide gel electrophoresis (Fig. 5A and B).

Short-strand RNA fragments inhibit HIV-1 replication

To test the inhibitory activity of the short-strand RNA fragments on HIV-1 replication in a transient assay, an HIV-1 expression plasmid (pNL4-3) and the short-strand RNA fragments were transfected into HeLa CD4+ cells. The virus production in the culture supernatant was monitored with the HIV-1 p24 antigen (HIV-1 gag gene product) assay. The results of the HIV-1 p24 assay indicated that these RNA fragments had anti-HIV activity and suppressed HIV-1 replication. Furthermore, our analysis revealed that the short-strand RNA fragments dose-dependently inhibited HIV-1 replication (Fig. 5C). These results suggest that the short-strand RNA fragments have a miRNA-like effect on the HIV-1 virus.

In conclusion, the short-strand RNA fragments (24 nt long) of R-Ψ-sgRNA cleaved using the Dicer enzyme exhibited anti-HIV-1 activity. The sequences of these RNA fragments were analyzed to determine the critical sequence for the anti-HIV-1 activity. These findings suggest that R-Ψ-sgRNA acts as a miRNA to inhibit HIV-1.

Acknowledgements

The authors thank Dr I.S.Y. Chen for providing pNL4-3lucΔenv and Makoto Abe and Yuji Yokota for their technical assistance. We also thank Dr Y. Habu for the valuable discussion. This study was supported in part by a Grant-in-Aid for AIDS research from the Ministry of Health, Labor, and Welfare, Japan, by a Grant from the Strategic Research Foundation Grant-aided Project for Private Universities from the Ministry of Education, Culture, Sport, Science and Technology, Japan (MEXT), and by a Grant-in-Aid for Science Research (C) from Japan Society for the Promotion of Science (JSPS), Japan.

Abbreviations:

HIV-1

human immunodeficiency virus type 1

miRNA

microRNA

nt

nucleotide

RISC

RNA-induced silencing complex

RNAi

RNA interference

sgRNA

small guide RNA

siRNA

short interfering RNA

tRNase ZL

tRNA 3′-processing endoribonuclease

Ψ

HIV-1 packaging signal

tRNAmet

methionine tRNA

Dicer

RNase III-family enzyme

References

1 

Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE and Mello CC: Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 39:806–811. 1998. View Article : Google Scholar

2 

Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K and Tuschl T: Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature. 411:494–498. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Sharp PA: RNA interference. Genes Dev. 15:485–490. 2001.PubMed/NCBI

4 

Hannon GJ: RNA interference. Nature. 418:244–251. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Coburn GA and Cullen BR: Potent and specific inhibition of human immunodeficiency virus type 1 replication by RNA interference. J Virol. 76:9225–9231. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Jacque JM, Trioques K and Stevenson M: Modulation of HIV-1 replication by RNA interference. Nature. 418:379–380. 2002. View Article : Google Scholar

7 

Lee NS, Dohjima T, Bauer G, Li H, Li MJ, Ehsani A, Salvaterra P and Rossi J: Expression of small interfering RNAs targeted against HIV-1 rev transcripts in human cells. Nat Biotech. 19:500–505. 2002.PubMed/NCBI

8 

Novina CD, Murray MF, Dykxhoorn DM, Beresford PJ, Riess J, Lee SK, Collman RG, Lieberman J, Shankar P and Sharp PA: siRNA-directed inhibition of HIV-1 infection. Nat Med. 8:681–686. 2002.PubMed/NCBI

9 

Yamamoto T, Omoto S, Mizuguchi M, Mizukami H, Okuyama H, Okada N, Saksena NK, Brisibe EA, Otake K and Fuji YR: Double-stranded nef RNA interferes with human immunodeficiency virus type 1 replication. Microbiol Immunol. 46:809–817. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Song E, Lee SK, Dykxhoorn DM, Novina C, Zhang D, Crawford K, Cerny J, Sharp PA, Lieberman J, Manjunath N and Shankar P: Sustained small interfering RNA-mediated human immunodeficiency virus type 1 inhibition in primary macrophages. J Virol. 77:7174–7181. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Park WS, Hayafune M, Miyano-Kurosaki N and Takaku H: Specific HIV-1 env gene silencing by small interfering RNAs in human peripheral blood mononuclear cells. Gene Ther. 10:2046–2050. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Martínez MA, Gutiérrez A, Armand-Ugón M, Blanco J, Parera M, Gómez J, Clotet B and Esté JA: Suppression of chemokine receptor expression by RNA interference allows for inhibition of HIV-1 replication. AIDS. 16:2385–2390. 2002.

13 

Qin XF, An DS, Chen IS and Baltimore D: Inhibiting HIV-1 infection in human T cells by lentiviral-mediated delivery of small interfering RNA against CCR5. Proc Natl Acad Sci USA. 100:183–188. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Anderson J, Banerjea A and Akkina R: Suppression of HIV-1 infection by a stem-loop structured anti-CXCR4 siRNA. AIDS Res Hum Retroviruses. 19:699–706. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Lee RC, Feinbaum RL and Ambros V: The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 75:843–854. 1993.

16 

Lagos-Quintana M, Rauhut R, Lendeckel W and Tuschl T: Identification of novel genes coding for small expressed RNAs. Science. 294:853–858. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Lau NC, Lim LP, Weinstein EG and Bartel DP: An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science. 294:858–862. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Bartel DP: MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Hammond SM: Dicing and slicing the core machinery of the RNA interference pathway. FEBS Lett. 579:5822–5829. 2005.PubMed/NCBI

20 

Tang G: siRNA and miRNA: an insight into RISCs. Trends Biochem Sci. 30:106–114. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Tomari Y and Zamore PD: Perspective: machines for RNAi. Genes Dev. 19:517–529. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Kim VN: MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol. 6:376–385. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Shiomi M: Functions and regulatory mechanisms of small RNAs expressed in living organisms. Exp Med. 25:794–799. 2007.

24 

Aravin AA, Lagos-Quintana M, Yalcin A, Zavolan M, Marks D, Snyder B, Gaasterland T, Meyer J and Tuschl T: The small RNA profile during Drosophila melanogaster development. Dev Cell. 5:337–350. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Aravin A, Gaidatzis D, Pfeffer S, Lagos-Quintana M, Landgraf P, Iovino N, Morris P, Brownstein MJ, Kuramochi-Miyagawa S, Nakano T, Chien M, Russo JJ, Ju J, Sheridan R, Sander C, Zavolan M and Tuschl T: A novel class of small RNAs bind to MILI protein in mouse testes. Nature. 442:203–207. 2006.PubMed/NCBI

26 

Girard A, Sachidanandam R, Hannon GJ and Carmell MA: A germline-specific class of small RNAs binds mammalian Piwi proteins. Nature. 442:199–202. 2006.PubMed/NCBI

27 

Grivna ST, Beyret E, Wang Z and Lin H: A novel class of small RNAs in mouse spermatogenic cells. Genes Dev. 20:1709–1714. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Lau NC, Seto AG, Kim J, Kuramochi-Miyagawa S, Nakano T, Bartel DP and Kingston RE: Characterization of the piRNA complex from rat testes. Science. 313:363–367. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Saito K, Nishida KM, Mori T, Kawamura Y, Miyoshi K, Nagami T, Siomi H and Siomi MC: Specific association of Piwi with rasiRNAs derived from retrotransposon and heterochromatic regions in the Drosophila genome. Genes Dev. 20:2214–2222. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Vagin VV, Sigova A, Li C, Seitz H, Gvozdev V and Zamore PD: A distinct small RNA pathway silences selfish genetic elements in the germline. Science. 313:320–324. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Watanabe T, Takeda A, Tsukiyama T, Mise K, Okuno T, Sasaki H, Minami N and Imai H: Identification and characterization of two novel classes of small RNAs in the mouse germline: retrotransposon-derived siRNAs in oocytes and germline small RNAs in testes. Genes Dev. 20:1732–1743. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Nishida KM, Saito K, Mori T, Kawamura Y, Nagami-Okada T, Inagaki S, Siomi H and Siomi MC: Gene silencing mechanisms mediated by Aubergine piRNA complexes in Drosophila male gonad. RNA. 13:1911–1922. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Klattenhoff C and Theurkauf W: Biogenesis and germline functions of piRNAs. Development. 135:3–9. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Kuramochi-Miyagawa S, Watanabe T, Gotoh K, Totoki Y, Toyoda A, Ikawa M, Asada N, Kojima K, Yamaguchi Y, Ijiri TW, Hata K, Li E, Matsuda Y, Kimura T, Okabe M, Sakaki Y, Sasaki H and Nakano T: DNA methylation of retrotransposon genes is regulated by Piwi family members MILI and MIWI2 in murine fetal testes. Genes Dev. 22:908–917. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Watanabe T, Totoki Y, Toyoda A, Kaneda M, Kuramochi-Miyagawa S, Obata Y, Chiba H, Kohara Y, Kono T, Nakano T, Surani MA, Sakaki Y and Sasaki H: Endogenous siRNAs from naturally formed dsRNAs regulate transcripts in mouse oocytes. Nature. 453:539–543. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Clever JL, Miranda D Jr and Parslow TG: RNA structure and packaging signals in the 5 leader region of the human immunodeficiency virus type 1 genome. J Virol. 76:12381–12387. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Russell RS, Hu J, Beriault V, Mouland AJ, Laughrea M, Kleiman L, Wainberg MA and Liang C: Sequences downstream of the 5 splice donor site are required for both packaging and dimerization of human immunodeficiency virus type 1 RNA. J Virol. 77:84–96. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Habu Y, Miyano-Kurosaki N, Kitano M, Endo Y, Yukita M, Ohira S and Takaku H, Nashimoto M and Takaku H: Inhibition of HIV-1 gene expression by retroviral vector-mediated small-guide RNAs that direct specific RNA cleavage by tRNase ZL. Nucleic Acids Res. 33:235–243. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Nashimoto M: Conversion of mammalian tRNA 3′ processing endoribonuclease to four-base-recognizing RNA cutters. Nucleic Acids Res. 23:3642–3647. 1995.

40 

Nashimoto M: Specific cleavage of target RNAs from HIV-1 with 5′ half tRNA by mammalian tRNA 3′ processing endoribonuclease. RNA. 2:523–524. 1996.

41 

Takaku H, Minagawa A, Takagi M and Nashimoto M: A candidate prostate cancer susceptibility gene encodes tRNA 3′ processing endoribonuclease. Nucleic Acids Res. 31:2272–2278. 2003.

42 

Takaku H, Minagawa A, Takagi M and Nashimoto M: The N-terminal half-domain of the long form of tRNase Z is required for the RNase 65 activity. Nucleic Acids Res. 32:4429–4438. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Kato K, Habu Y and Takaku H: Micro RNA like inhibition of HIV-1 replication. Nucleic Acids Symp Ser (Oxf). 52:509–510. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Sugiyama R, Hayafune M, Habu Y, Yamamoto N and Takaku H: HIV-1 RT-dependent DNAzyme expression inhibits HIV-1 replication without the emergence of escape viruses. Nucleic Acids Res. 39:589–598. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Keith G, Heitzler J, el Adlouni C, Glasser AL, Fix C, Desgres J and Dirheimer G: The primary structure of cytoplasmic initiator tRNA(Met) from Schizosaccharomyces pombe. Nucleic Acids Res. 21:29491993. View Article : Google Scholar : PubMed/NCBI

46 

Planelles V, Bachelerie F, Jowett JB, Haislip A, Xie Y, Banooni P, Masuda T and Chen IS: Fate of the human immunodeficiency virus type 1 provirus in infected cells: a role for vpr. J Virol. 69:5883–5889. 1995.PubMed/NCBI

47 

Sakai A, Hirabayashi Y, Aizawa S, Tanaka M, Ida S and Oka S: Investigation of a new p24 antigen detection system by the chemiluminescence-enzyme-immuno-assay. Kansenshogaku Zasshi. 73:205–212. 1999.(In Japanese).

48 

Zuker M: Mfold web server for nucleic acid folding and hybridization prediction. Nucleic Acids Res. 31:3406–3415. 2003. View Article : Google Scholar : PubMed/NCBI

49 

Enright AJ, John B, Tushl T, Sander C and Marks DS: Micro RNA targets in Drosophila. Genom Biol. 5:R12003. View Article : Google Scholar : PubMed/NCBI

50 

Myers JW, Jones JT, Meyer T and Ferrell JE Jr: Recombinant Dicer efficiently converts large dsRNAs into siRNAs suitable for gene silencing. Nat Biotechnol. 3:324–328. 2003. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January 2013
Volume 31 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kato K, Senoki T and Takaku H: Inhibition of HIV-1 replication by RNA with a microRNA-like function. Int J Mol Med 31: 252-258, 2013
APA
Kato, K., Senoki, T., & Takaku, H. (2013). Inhibition of HIV-1 replication by RNA with a microRNA-like function. International Journal of Molecular Medicine, 31, 252-258. https://doi.org/10.3892/ijmm.2012.1170
MLA
Kato, K., Senoki, T., Takaku, H."Inhibition of HIV-1 replication by RNA with a microRNA-like function". International Journal of Molecular Medicine 31.1 (2013): 252-258.
Chicago
Kato, K., Senoki, T., Takaku, H."Inhibition of HIV-1 replication by RNA with a microRNA-like function". International Journal of Molecular Medicine 31, no. 1 (2013): 252-258. https://doi.org/10.3892/ijmm.2012.1170