Summary

Evaluering af effekt og toksicitet af RNA Målretning HIV-1 Produktion til brug i Gene eller Drug Therapy

Published: September 05, 2016
doi:

Summary

Methods to evaluate the efficacy and toxicity of RNA molecules targeting post-integration steps of the HIV-1 replication cycle are described. These methods are useful for screening new molecules and optimizing the format of existing ones.

Abstract

Small RNA therapies targeting post-integration steps in the HIV-1 replication cycle are among the top candidates for gene therapy and have the potential to be used as drug therapies for HIV-1 infection. Post-integration inhibitors include ribozymes, short hairpin (sh) RNAs, small interfering (si) RNAs, U1 interference (U1i) RNAs and RNA aptamers. Many of these have been identified using transient co-transfection assays with an HIV-1 expression plasmid and some have advanced to clinical trials. In addition to measures of efficacy, small RNAs have been evaluated for their potential to affect the expression of human RNAs, alter cell growth and/or differentiation, and elicit innate immune responses. In the protocols described here, a set of transient transfection assays designed to evaluate the efficacy and toxicity of RNA molecules targeting post-integration steps in the HIV-1 replication cycle are described. We have used these assays to identify new ribozymes and optimize the format of shRNAs and siRNAs targeting HIV-1 RNA. The methods provide a quick set of assays that are useful for screening new anti-HIV-1 RNAs and could be adapted to screen other post-integration inhibitors of HIV-1 replication.

Introduction

En begrænsning ved nuværende HIV-1 behandlinger er, at de skal være kronisk administreret for at forhindre progression af sygdommen. Transplantation af HIV-1 resistent T-lymfocyt, eller hæmatopoietiske stamceller, har potentialet til at give langsigtet kontrol af HIV-1-replikation i fravær af lægemiddelbehandling 1,2 og kan også være en effektiv tilgang til at opnå en HIV-1 kur 3. En måde at gøre celler modstandsdygtige mod HIV-1-replikation er at indsætte et eller flere gener der koder for anti-HIV-1 RNA'er eller peptider i en inficeret individs celler under en autolog transplantation 4. Adskillige kandidat anti-HIV-1-gener er blevet designet med nogle ind kliniske forsøg i kombinationer af to 5 eller tre 6, for at forhindre udviklingen af HIV-1 resistens over for en enkelt gen.

Anti-HIV-1 RNA er blandt de bedste kandidater til kombination genterapi på grund af deres ringe potentiale til at fremkalde immunresponser og fordi detranskriberes fra meget korte gensekvenser. Nogle anti-HIV-1 RNA er designet til at målrette viral indtrængen og integration. Men de fleste anti-HIV-1 RNA målrette post-integration trin i den virale livscyklus (figur 1). Post-integration hæmmere omfatter lokkefugle RNA, rettet mod de HIV-1 regulerende proteiner Tat eller Rev 1, og antisense-baserede RNA, rettet mod forskellige steder i HIV-1 RNA, såsom ribozymer 7, shRNAs 8 og U1i RNA 9. Metoder, der er anvendt til at sammenligne virkningen af anti-HIV-1 RNA inkluderer monitorering viral replikation i celler transduceret med gener, der koder for kandidat RNA'er og måle viral produktion i celler transient transficeret med plasmider, der udtrykker kandidat RNA'er og en HIV-1 ekspressionsplasmid 10 -13. Vi har tidligere anvendt et HIV-1-produktion assay til screening HIV-1 RNA til nye ribozym målsteder 13-15. Disse fremgangsmåder er siden blevet raffineret for at optimere udformningen af ​​et RNAinterferens molekyle udtrykt fra plasmid-DNA som en shRNA eller leveres som et syntetisk siRNA 16. Assayet måler produktionen af modne virus fra humane embryoniske nyre (HEK) 293T-celler, og kan anvendes til at sammenligne virkningerne af inhibitorer, der er målrettet efter integration trin i HIV-1-replikation cyklus (figur 1). For inhibitorer, der er målrettet præ-integration trin, er alternative assays, såsom et TZM-bl celle infektivitet assay 17 er nødvendige til vurdering antiviral virkningsfuldhed.

Større sikkerhedsproblemer for levering af anti-HIV-1 RNA i klinikken omfatter potentielle off-target effekter på humane RNA eller proteiner og aktivering af medfødte immun sensorer. For at evaluere toksiciteten af anti-HIV-1 siRNAs, har vi anvendt en celleviabilitetstest i forskellige cellelinier 16. Vi målte også aktivering af det dobbeltstrengede RNA immune sensorer, RNA aktiveret protein kinase R (PKR) og Toll like receptor 3 (TLR3), samt expression af interferon stimulerede gen, ADAR1 P150. Disse assays kan anvendes til at bekræfte, at effektiviteten af ​​anti-HIV-1 RNA'er ikke skyldes indirekte virkninger på cellernes levedygtighed eller immun sensor aktivering. De er også nyttige i at udelukke kandidat RNA'er med potentielle toksiciteter fra yderligere udvikling.

I de følgende protokoller, procedurer identificere nye terapeutiske RNA og optimere format eksisterende beskrives. Fremgangsmåderne er nyttige til screening RNA baserede post-integration inhibitorer af HIV-1 replikation og kan tilpasses til at screene andre post-integration inhibitorer, såsom små molekyler rettet Rev medieret eksport af viralt RNA 18 eller CRISPR / Cas systemer designet til at målrette integreret HIV-1-DNA 19.

Protocol

1. Celler og transfektioner Kultur HEK 293T celler i Dulbeccos modificerede Eagle-medium (DMEM) suppleret med 10% føtalt bovint serum (FBS) og 1% penicillin / streptomycin. En 2 x 10 5 celler / ml suspension i celledyrkningsmediet. Der tilsættes 500, 100 og 1000 pi af cellesuspensionen til hver brønd i 24-brønds, 96-brønds og 12-brønds plader, til virusproduktion, cellelevedygtighed og immunaktivering assays, henholdsvis (figur 2A). Vend forsigtigt pladerne og inkub…

Representative Results

En generel skematisk af procedurerne er vist i figur 2 med et eksempel transfektion plan for tre test RNA'er og en kontrol-RNA tilvejebragt i figur 2B. For virale produktion og cellelevedygtighedsassays, udlæsningen for hver test-konstruktionen er normaliseret til en negativ kontrol. Replikater transficeres i sæt, således at hver test RNA normaliseret til dens tilstødende negativ kontrol. Dette gøres for at undgå ukorrekte oplysninger relateret…

Discussion

HIV-1-produktion assayet beskrevet blev udført under anvendelse HEK293T celler (figur 2) og svarer til assays anvendes til at screene HIV-1 RNA til effektiv ribozym 13, shRNA 10,29, siRNA 30, og U1i RNA 11,31 målsteder. Hjælp af forskellige metoder til at kvantificere HIV-1-produktion, har de fleste undersøgelser målte virusproduktion 48 timer efter co-transfektion af en HIV-1-ekspressionsplasmid med kandidat- RNA'er. Efter produktionen af ​​HIV-1…

Disclosures

The authors have nothing to disclose.

Acknowledgements

Arbejdet præsenteres her blev støttet af den canadiske Institutes of Health Research (CIHR) (giver DCB-120.266, PPP-133.377 og HBF-348.967 til AG).

Materials

DMEM HyClone GE Healthcare SH30243.01
FBS HyClone GE Healthcare SH30396.03
Penicillin/Streptomycin Gibco Thermo Fisher 15140-122
Cell culture plates, 96 well, 24 well, 6 well. Corning 353075, 353047, 353043
Micro tubes Axygen Corning 311-08-051
Low molecular weight Poly I:C InvivoGen 3182-29-6
DharmaFECT-1 Dharmacon T-2001-01 transfection reagent for synthetic RNAs
TransIT-LT1 Mirus MIR 2300 transfection reagent for RNA expression plasmids
Nonidet P40 (NP-40) USB 19628
[32P]dTTP Perkin Elmer BLU505H
poly(A) RNA template  Sigma-Aldrich 10108626001
oligo(dT)12-18 DNA primer Thermo Fisher 18418-012
DEAE filtermat paper  Perkin Elmer 1450-522
Microplate scintillation counter Perkin Elmer 1450-024
MTT Sigma-Aldrich M-2128
DPBS HyClone GE Healthcare SH30028.02
Microplate spectrophotometer Bio-rad 1706930
Lysis buffer tablets Roche 4693159001, 4906837001 protease and phosphatase inhibitors
Microcentrifuge Eppendorf 5415R
Bradford reagent Bio-rad 500-0006
Gel running chamber Hoefer SE600
Semi-dry transfer cell Bio-rad 1703940
Protein ladder EZ-Run Thermo Fisher BP3603-500
Nitrocellulose membrane Bio-rad 162-0094
BSA Sigma-Aldrich A9647-1006
Antibody stripping solution Millipore 2504
ECL – Pierce Thermo Fisher  PI32106
ADAR1 antibody from Dr. B.L. Bass
phospho-T446-PKR antibody Abcam ab32036
phospho-S396-IRF3 antibody Cell Signaling 4947
PKR antibody from Dr. A. Hovanessian
IRF3 antibody Cell Signaling 11904
Actin antibody Millipore MAB1501
Peroxidase-labeled goat anti-rabbit KPL 474-1506
Peroxidase-labeled goat anti-mouse KPL 474-1806
Ponceau S  Sigma-Aldrich 6226-79-5

References

  1. Hoxie, J. A., June, C. H. Novel cell and gene therapies for HIV. Cold Spring Harb Perspect Med. 2, a007179 (2012).
  2. Bobbin, M. L., Burnett, J. C., Rossi, J. J. RNA interference approaches for treatment of HIV-1 infection. Genome Med. 7, 50 (2015).
  3. Allers, K., et al. Evidence for the cure of HIV infection by CCR5Delta32/Delta32 stem cell transplantation. Blood. 117, 2791-2799 (2011).
  4. DiGiusto, D. L., et al. Development of hematopoietic stem cell based gene therapy for HIV-1 infection: considerations for proof of concept studies and translation to standard medical practice. Viruses. 5, 2898-2919 (2013).
  5. Burke, B. P., et al. Engineering Cellular Resistance to HIV-1 Infection In Vivo Using a Dual Therapeutic Lentiviral Vector. Mol Ther Nucleic Acids. 4, e236 (2015).
  6. DiGiusto, D. L., et al. RNA-based gene therapy for HIV with lentiviral vector-modified CD34(+) cells in patients undergoing transplantation for AIDS-related lymphoma. Sci Transl Med. 2, 36ra43 (2010).
  7. Scarborough, R. J., Gatignol, A. HIV and Ribozymes. Adv Exp Med Biol. 848, 97-116 (2015).
  8. Eekels, J. J., Berkhout, B. Toward a durable treatment of HIV-1 infection using RNA interference. Prog Mol Biol Transl Sci. 102, 141-163 (2011).
  9. Blazquez, L., Fortes, P. U1 interference (U1i) for Antiviral Approaches. Adv Exp Med Biol. 848, 51-69 (2015).
  10. McIntyre, G. J., et al. 96 shRNAs designed for maximal coverage of HIV-1 variants. Retrovirology. 6, 55 (2009).
  11. Sajic, R., et al. Use of modified U1 snRNAs to inhibit HIV-1 replication. Nucleic Acids Res. 35, 247-255 (2007).
  12. Low, J. T., et al. SHAPE-directed discovery of potent shRNA inhibitors of HIV-1. Mol Ther. 20, 820-828 (2012).
  13. Scarborough, R. J., et al. A Conserved Target Site in HIV-1 Gag RNA is Accessible to Inhibition by Both an HDV Ribozyme and a Short Hairpin RNA. Mol Ther Nucleic Acids. 3, e178 (2014).
  14. Lainé, S., et al. In vitro and in vivo cleavage of HIV-1 RNA by new SOFA-HDV ribozymes and their potential to inhibit viral replication. RNA Biol. 8, 343-353 (2011).
  15. Scarborough, R. J., Lévesque, M. V., Perreault, J. P., Gatignol, A. Design and Evaluation of Clinically Relevant SOFA-HDV Ribozymes Targeting HIV RNA. Methods Mol Biol. 1103, 31-43 (2014).
  16. Scarborough, R. J., Adams, K. L., Daher, A., Gatignol, A. Effective Inhibition of HIV-1 Production by Short Hairpin RNAs and Small Interfering RNAs Targeting a Highly Conserved Site in HIV-1 Gag RNA Is Optimized by Evaluating Alternative Length Formats. Antimicrob Agents Chemother. 59, 5297-5305 (2015).
  17. Sarzotti-Kelsoe, M., et al. Optimization and validation of the TZM-bl assay for standardized assessments of neutralizing antibodies against HIV-1. J Immunol Methods. 409, 131-146 (2014).
  18. Campos, N., et al. Long lasting control of viral rebound with a new drug ABX464 targeting Rev – mediated viral RNA biogenesis. Retrovirology. 12, 1-15 (2015).
  19. Zhu, W., et al. The CRISPR/Cas9 system inactivates latent HIV-1 proviral DNA. Retrovirology. 12, 22 (2015).
  20. Bounou, S., Leclerc, J. E., Tremblay, M. J. Presence of host ICAM-1 in laboratory and clinical strains of human immunodeficiency virus type 1 increases virus infectivity and CD4(+)-T-cell depletion in human lymphoid tissue, a major site of replication in vivo. J Virol. 76, 1004-1014 (2002).
  21. Shan, L., et al. A novel PCR assay for quantification of HIV-1 RNA. J Virol. 87, 6521-6525 (2013).
  22. Clerzius, G., et al. The PKR activator, PACT, becomes a PKR inhibitor during HIV-1 replication. Retrovirology. 10, 96 (2013).
  23. Bradford, M. M. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 72, 248-254 (1976).
  24. Stoscheck, C. M. Quantitation of protein. Methods Enzymol. 182, 50-68 (1990).
  25. Battisti, P. L., et al. Additive activity between the trans-activation response RNA-binding protein, TRBP2, and cyclin T1 on HIV type 1 expression and viral production in murine cells. AIDS Res Hum Retroviruses. 19, 767-778 (2003).
  26. Bannwarth, S., et al. Cell-specific regulation of TRBP1 promoter by NF-Y transcription factor in lymphocytes and astrocytes. J Mol Biol. 355, 898-910 (2006).
  27. Clerzius, G., et al. ADAR1 interacts with PKR during human immunodeficiency virus infection of lymphocytes and contributes to viral replication. J Virol. 83, 10119-10128 (2009).
  28. Burugu, S., Daher, A., Meurs, E. F., Gatignol, A. HIV-1 translation and its regulation by cellular factors PKR and PACT. Virus Res. 193, 65-77 (2014).
  29. ter Brake, O., Konstantinova, P., Ceylan, M., Berkhout, B. Silencing of HIV-1 with RNA interference: a multiple shRNA approach. Mol Ther. 14, 883-892 (2006).
  30. Naito, Y., et al. Optimal design and validation of antiviral siRNA for targeting HIV-1. Retrovirology. 4, 80 (2007).
  31. Mandal, D., Feng, Z., Stoltzfus, C. M. Excessive RNA splicing and inhibition of HIV-1 replication induced by modified U1 small nuclear RNAs. J Virol. 84, 12790-12800 (2010).
  32. Chang, Z., et al. Enhanced expression and HIV-1 inhibition of chimeric tRNA(Lys3)-ribozymes under dual U6 snRNA and tRNA promoters. Mol Ther. 6, 481-489 (2002).
  33. Chang, L. J., Liu, X., He, J. Lentiviral siRNAs targeting multiple highly conserved RNA sequences of human immunodeficiency virus type 1. Gene Ther. 12, 1133-1144 (2005).
  34. Daniels, S. M., et al. HIV-1 RRE RNA acts as an RNA silencing suppressor by competing with TRBP-bound siRNAs. RNA Biol. 12, 123-135 (2015).
  35. Castanotto, D., et al. Combinatorial delivery of small interfering RNAs reduces RNAi efficacy by selective incorporation into RISC. Nucleic Acids Res. 35, 5154-5164 (2007).
  36. Vickers, T. A., Sabripour, M., Crooke, S. T. U1 adaptors result in reduction of multiple pre-mRNA species principally by sequestering U1snRNP. Nucleic Acids Res. 39, e71 (2011).
  37. Tai, C. J., Li, C. L., Tai, C. J., Wang, C. K., Lin, L. T. Early Viral Entry Assays for the Identification and Evaluation of Antiviral Compounds. J Vis Exp. , (2015).
  38. Riss, T. L., et al., Sittampalam, G. S., et al. . Assay Guidance Manual. , (2004).
  39. Reynolds, A., et al. Induction of the interferon response by siRNA is cell type- and duplex length-dependent. Rna. 12, 988-993 (2006).
  40. Whitehead, K. A., Dahlman, J. E., Langer, R. S., Anderson, D. G. Silencing or stimulation? siRNA delivery and the immune system. Annu Rev Chem Biomol Eng. 2, 77-96 (2011).
check_url/54486?article_type=t

Play Video

Cite This Article
Scarborough, R. J., Adams, K. L., Del Corpo, O., Daher, A., Gatignol, A. Evaluation of the Efficacy And Toxicity of RNAs Targeting HIV-1 Production for Use in Gene or Drug Therapy. J. Vis. Exp. (115), e54486, doi:10.3791/54486 (2016).

View Video