#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Highly Significant Antiviral Activity of HIV-1 LTR-Specific Tre-Recombinase in Humanized Mice


Stable integration of HIV proviral DNA into host cell chromosomes, a hallmark and essential feature of the retroviral life cycle, establishes the infection permanently. Current antiretroviral combination drug therapy cannot cure HIV infection. However, expressing an engineered HIV-1 long terminal repeat (LTR) site-specific recombinase (Tre), shown to excise integrated proviral DNA in vitro, may provide a novel and highly promising antiviral strategy. We report here the conditional expression of Tre-recombinase from an advanced lentiviral self-inactivation (SIN) vector in HIV-infected cells. We demonstrate faithful transgene expression, resulting in accurate provirus excision in the absence of cytopathic effects. Moreover, pronounced Tre-mediated antiviral effects are demonstrated in vivo, particularly in humanized Rag2−/−γc−/− mice engrafted with either Tre-transduced primary CD4+ T cells, or Tre-transduced CD34+ hematopoietic stem and progenitor cells (HSC). Taken together, our data support the use of Tre-recombinase in novel therapy strategies aiming to provide a cure for HIV.


Vyšlo v časopise: Highly Significant Antiviral Activity of HIV-1 LTR-Specific Tre-Recombinase in Humanized Mice. PLoS Pathog 9(9): e32767. doi:10.1371/journal.ppat.1003587
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003587

Souhrn

Stable integration of HIV proviral DNA into host cell chromosomes, a hallmark and essential feature of the retroviral life cycle, establishes the infection permanently. Current antiretroviral combination drug therapy cannot cure HIV infection. However, expressing an engineered HIV-1 long terminal repeat (LTR) site-specific recombinase (Tre), shown to excise integrated proviral DNA in vitro, may provide a novel and highly promising antiviral strategy. We report here the conditional expression of Tre-recombinase from an advanced lentiviral self-inactivation (SIN) vector in HIV-infected cells. We demonstrate faithful transgene expression, resulting in accurate provirus excision in the absence of cytopathic effects. Moreover, pronounced Tre-mediated antiviral effects are demonstrated in vivo, particularly in humanized Rag2−/−γc−/− mice engrafted with either Tre-transduced primary CD4+ T cells, or Tre-transduced CD34+ hematopoietic stem and progenitor cells (HSC). Taken together, our data support the use of Tre-recombinase in novel therapy strategies aiming to provide a cure for HIV.


Zdroje

1. ThompsonMA, AbergJA, CahnP, MontanerJS, RizzardiniG, et al. (2010) Antiretroviral treatment of adult HIV infection: 2010 recommendations of the International AIDS Society-USA panel. JAMA 304: 321–333.

2. SchackmanBR, GeboKA, WalenskyRP, LosinaE, MuccioT, et al. (2006) The lifetime cost of current human immunodeficiency virus care in the United States. Med Care 44: 990–997.

3. ChenRY, AccorttNA, WestfallAO, MugaveroMJ, RaperJL, et al. (2006) Distribution of health care expenditures for HIV-infected patients. Clin Infect Dis 42: 1003–1010.

4. DeeksSG, PhillipsAN (2009) HIV infection, antiretroviral treatment, ageing, and non-AIDS related morbidity. BMJ 338: a3172.

5. CalmyA, HirschelB, CooperDA, CarrA (2009) A new era of antiretroviral drug toxicity. Antivir Ther 14: 165–179.

6. DeeksSG (2011) HIV infection, inflammation, immunosenescence, and aging. Annu Rev Med 62: 141–155.

7. LewinSR, EvansVA, ElliottJH, SpireB, ChomontN (2011) Finding a cure for HIV: will it ever be achievable? J Int AIDS Soc 14: 4.

8. Schulze zur WieschJ, van LunzenJ (2011) Hide and seek… Can we eradicate HIV by treatment intensification? J Infect Dis 203: 894–897.

9. RichmanDD, MargolisDM, DelaneyM, GreeneWC, HazudaD, et al. (2009) The challenge of finding a cure for HIV infection. Science 323: 1304–1307.

10. Iglesias-UsselMD, RomerioF (2011) HIV reservoirs: the new frontier. AIDS Rev 13: 13–29.

11. LafeuilladeA, StevensonM (2011) The search for a cure for persistent HIV reservoirs. AIDS Rev 13: 63–66.

12. MargolisDM (2011) Eradication therapies for HIV Infection: time to begin again. AIDS Res Hum Retroviruses 27: 347–353.

13. SmithMZ, WightmanF, LewinSR (2012) HIV Reservoirs and Strategies for Eradication. Curr HIV/AIDS Rep 9: 5–15.

14. LafeuilladeA (2012) Eliminating the HIV Reservoir. Curr HIV/AIDS Rep 9: 121–131.

15. DurandCM, BlanksonJN, SilicianoRF (2012) Developing strategies for HIV-1 eradication. Trends Immunol 33: 554–562.

16. ChoudharySK, MargolisDM (2011) Curing HIV: Pharmacologic approaches to target HIV-1 latency. Annu Rev Pharmacol Toxicol 51: 397–418.

17. WightmanF, EllenbergP, ChurchillM, LewinSR (2012) HDAC inhibitors in HIV. Immunol Cell Biol 90: 47–54.

18. SarkarI, HauberI, HauberJ, BuchholzF (2007) HIV-1 proviral DNA excision using an evolved recombinase. Science 316: 1912–1915.

19. BuchholzF, HauberJ (2011) In vitro evolution and analysis of HIV-1 LTR-specific recombinases. Methods 53: 102–109.

20. BuchholzF, HauberJ (2013) Engineered DNA modifying enzymes: components of a future strategy to cure HIV/AIDS. Antiviral Res 97: 211–217.

21. BlackardJT, RenjifoBR, MwakagileD, MontanoMA, FawziWW, et al. (1999) Transmission of human immunodeficiency type 1 viruses with intersubtype recombinant long terminal repeat sequences. Virology 254: 220–225.

22. van LunzenJ, FehseB, HauberJ (2011) Gene Therapy Strategies: Can We Eradicate HIV? Curr HIV/AIDS Rep 8: 78–84.

23. RossiJJ, JuneCH, KohnDB (2007) Genetic therapies against HIV. Nat Biotechnol 25: 1444–1454.

24. SchererLJ, RossiJJ (2011) Ex vivo gene therapy for HIV-1 treatment. Hum Mol Genet 20: R100–R107.

25. KiemHP, JeromeKR, DeeksSG, McCuneJM (2012) Hematopoietic-stem-cell-based gene therapy for HIV disease. Cell Stem Cell 10: 137–147.

26. BaltimoreD (1988) Gene therapy. Intracellular immunization. Nature 335: 395–396.

27. DullT, ZuffereyR, KellyM, MandelRJ, NguyenM, et al. (1998) A third-generation lentivirus vector with a conditional packaging system. J Virol 72: 8463–8471.

28. SchambachA, GallaM, MaetzigT, LoewR, BaumC (2007) Improving transcriptional termination of self-inactivating gamma-retroviral and lentiviral vectors. Mol Ther 15: 1167–1173.

29. EmermanM, MalimMH (1998) HIV-1 regulatory/accessory genes: keys to unraveling viral and host cell biology. Science 280: 1880–1884.

30. LarochelleN, StuckaR, RiegerN, SchermellehL, SchiednerG, et al. (2011) Genomic integration of adenoviral gene transfer vectors following transduction of fertilized mouse oocytes. Transgenic Res 20: 123–135.

31. ParuzynskiA, ArensA, GabrielR, BartholomaeCC, ScholzS, et al. (2010) Genome-wide high-throughput integrome analyses by nrLAM-PCR and next-generation sequencing. Nat Protoc 5: 1379–1395.

32. SchröckE, ZschieschangP, O'BrienP, HelmrichA, HardtT, et al. (2006) Spectral karyotyping of human, mouse, rat and ape chromosomes–applications for genetic diagnostics and research. Cytogenet Genome Res 114: 199–221.

33. SchröckE, WeaverZ, AlbertsonD (2001) Comparative genomic hybridization (CGH)–detection of unbalanced genetic aberrations using conventional and micro-array techniques. Curr Protoc Cytom Chapter 8: Unit 8.12.

34. MariyannaL, PriyadarshiniP, Hofmann-SieberH, KrepstakiesM, WalzN, et al. (2012) Excision of HIV-1 Proviral DNA by Recombinant Cell Permeable Tre-Recombinase. PLoS One 7: e31576.

35. SurendranathV, ChusainowJ, HauberJ, BuchholzF, HabermannBH (2010) SeLOX–a locus of recombination site search tool for the detection and directed evolution of site-specific recombination systems. Nucleic Acids Res 38: W293–W298.

36. MazurierF, FontanellasA, SalesseS, TaineL, LandriauS, et al. (1999) A novel immunodeficient mouse model–RAG2 x common cytokine receptor gamma chain double mutants–requiring exogenous cytokine administration for human hematopoietic stem cell engraftment. J Interferon Cytokine Res 19: 533–541.

37. TraggiaiE, ChichaL, MazzucchelliL, BronzL, PiffarettiJC, et al. (2004) Development of a human adaptive immune system in cord blood cell-transplanted mice. Science 304: 104–107.

38. BaenzigerS, TussiwandR, SchlaepferE, MazzucchelliL, HeikenwalderM, et al. (2006) Disseminated and sustained HIV infection in CD34+ cord blood cell-transplanted Rag2−/−gamma c−/− mice. Proc Natl Acad Sci U S A 103: 15951–15956.

39. NeaguMR, ZieglerP, PertelT, Strambio-De-CastilliaC, GrutterC, et al. (2009) Potent inhibition of HIV-1 by TRIM5-cyclophilin fusion proteins engineered from human components. J Clin Invest 119: 3035–3047.

40. BergesBK, RowanMR (2011) The utility of the new generation of humanized mice to study HIV-1 infection: transmission, prevention, pathogenesis, and treatment. Retrovirology 8: 65.

41. ZouW, DentonPW, WatkinsRL, KriskoJF, NochiT, et al. (2012) Nef functions in BLT mice to enhance HIV-1 replication and deplete CD4+CD8+ thymocytes. Retrovirology 9: 44.

42. DiGiustoDL, KrishnanA, LiL, LiH, LiS, et al. (2010) 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.

43. NeffCP, ZhouJ, RemlingL, KuruvillaJ, ZhangJ, et al. (2011) An aptamer-siRNA chimera suppresses HIV-1 viral loads and protects from helper CD4(+) T cell decline in humanized mice. Sci Transl Med 3: 66ra6.

44. ter BrakeO, LegrandN, von EijeKJ, CentlivreM, SpitsH, et al. (2009) Evaluation of safety and efficacy of RNAi against HIV-1 in the human immune system (Rag-2(−/−)gammac(−/−)) mouse model. Gene Ther 16: 148–153.

45. van LunzenJ, GlaunsingerT, StahmerI, von BaehrV, BaumC, et al. (2007) Transfer of autologous gene-modified T cells in HIV-infected patients with advanced immunodeficiency and drug-resistant virus. Mol Ther 15: 1024–1033.

46. KimpelJ, BraunSE, QiuG, WongFE, ConolleM, et al. (2010) Survival of the fittest: positive selection of CD4+ T cells expressing a membrane-bound fusion inhibitor following HIV-1 infection. PLoS One 5: e12357.

47. HütterG, NowakD, MossnerM, GanepolaS, MüßigA, et al. (2009) Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med 360: 692–698.

48. AllersK, HutterG, HofmannJ, LoddenkemperC, RiegerK, et al. (2011) Evidence for the cure of HIV infection by CCR5Delta32/Delta32 stem cell transplantation. Blood 117: 2791–2799.

49. PerezEE, WangJ, MillerJC, JouvenotY, KimKA, et al. (2008) Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol 26: 808–816.

50. HoltN, WangJ, KimK, FriedmanG, WangX, et al. (2010) Human hematopoietic stem/progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HIV-1 in vivo. Nat Biotechnol 28: 839–847.

51. KatlamaC, DeeksSG, AutranB, Martinez-PicadoJ, van LunzenJ, et al. (2013) Barriers to a cure for HIV: new ways to target and eradicate HIV-1 reservoirs. Lancet 381: 2109–2117.

52. BartonKM, BurchBD, Soriano-SarabiaN, MargolisDM (2013) Prospects for treatment of latent HIV. Clin Pharmacol Ther 93: 46–56.

53. MarsdenMD, ZackJA (2013) HIV/AIDS eradication. Bioorg Med Chem Lett 23: 4003–4010.

54. ShanL, SilicianoRF (2013) From reactivation of latent HIV-1 to elimination of the latent reservoir: The presence of multiple barriers to viral eradication. Bioessays 35: 544–552.

55. ArchinNM, EspesethA, ParkerD, CheemaM, HazudaD, et al. (2009) Expression of latent HIV induced by the potent HDAC inhibitor suberoylanilide hydroxamic acid. AIDS Res Hum Retroviruses 25: 207–212.

56. ArchinNM, LibertyAL, KashubaAD, ChoudharySK, KurucJD, et al. (2012) Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature 487: 482–485.

57. ShanL, DengK, ShroffNS, DurandCM, RabiSA, et al. (2012) Stimulation of HIV-1-specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation. Immunity 36: 491–501.

58. DeeksSG, McCuneJM (2010) Can HIV be cured with stem cell therapy? Nat Biotechnol 28: 807–810.

59. ZintzarasE, KowaldA (2011) A mathematical model of HIV dynamics in the presence of a rescuing virus with replication deficiency. Theory Biosci 130: 127–134.

60. LachmannN, BrennigS, PfaffN, SchermeierH, DahlmannJ, et al. (2012) Efficient in vivo regulation of cytidine deaminase expression in the haematopoietic system using a doxycycline-inducible lentiviral vector system. Gene Ther 20: 298–307.

61. LudwigC, WagnerR (2007) Virus-like particles-universal molecular toolboxes. Curr Opin Biotechnol 18: 537–545.

62. Van DuyneGD (2001) A structural view of cre-loxp site-specific recombination. Annu Rev Biophys Biomol Struct 30: 87–104.

63. PattanayakV, RamirezCL, JoungJK, LiuDR (2011) Revealing off-target cleavage specificities of zinc-finger nucleases by in vitro selection. Nat Methods 8: 765–770.

64. GabrielR, LombardoA, ArensA, MillerJC, GenoveseP, et al. (2011) An unbiased genome-wide analysis of zinc-finger nuclease specificity. Nat Biotechnol 29: 816–823.

65. McIntyreGJ, GronemanJL, YuYH, JaramilloA, ShenS, et al. (2009) 96 shRNAs designed for maximal coverage of HIV-1 variants. Retrovirology 6: 55.

66. SchambachA, BohneJ, ChandraS, WillE, MargisonGP, et al. (2006) Equal potency of gammaretroviral and lentiviral SIN vectors for expression of O6-methylguanine-DNA methyltransferase in hematopoietic cells. Mol Ther 13: 391–400.

67. EgelhoferM, BrandenburgG, MartiniusH, Schult-DietrichP, MelikyanG, et al. (2004) Inhibition of human immunodeficiency virus type 1 entry in cells expressing gp41-derived peptides. J Virol 78: 568–575.

68. SchambachA, GallaM, ModlichU, WillE, ChandraS, et al. (2006) Lentiviral vectors pseudotyped with murine ecotropic envelope: increased biosafety and convenience in preclinical research. Exp Hematol 34: 588–592.

69. WeberK, MockU, PetrowitzB, BartschU, FehseB (2010) Lentiviral gene ontology (LeGO) vectors equipped with novel drug-selectable fluorescent proteins: new building blocks for cell marking and multi-gene analysis. Gene Ther 17: 511–520.

70. HwangSS, BoyleTJ, LyerlyHK, CullenBR (1991) Identification of the envelope V3 loop as the primary determinant of cell tropism in HIV-1. Science 253: 71–74.

71. ShanerNC, CampbellRE, SteinbachPA, GiepmansBN, PalmerAE, et al. (2004) Improved monomeric red, orange and yellow fluorescent proteins derived from Discosoma sp. red fluorescent protein. Nat Biotechnol 22: 1567–1572.

72. BeyerWR, WestphalM, OstertagW, von LaerD (2002) Oncoretrovirus and lentivirus vectors pseudotyped with lymphocytic choriomeningitis virus glycoprotein: generation, concentration, and broad host range. J Virol 76: 1488–1495.

73. ChemnitzJ, PieperD, GrüttnerC, HauberJ (2009) Phosphorylation of the HuR ligand APRIL by casein kinase 2 regulates CD83 expression. Eur J Immunol 39: 267–279.

74. FriesB, HeukeshovenJ, HauberI, GrüttnerC, StockingC, et al. (2007) Analysis of Nucleocytoplasmic Trafficking of the HuR Ligand APRIL and Its Influence on CD83 Expression. J Biol Chem 282: 4504–4515.

75. Schulze zur WieschJ, ThomssenA, HartjenP, TothI, LehmannC, et al. (2011) Comprehensive analysis of frequency and phenotype of T regulatory cells in HIV infection: CD39 expression of FoxP3+ T regulatory cells correlates with progressive disease. J Virol 85: 1287–1297.

76. Schulze zur WieschJ, PieperD, StahmerI, EiermannT, BuggischP, et al. (2009) Sustained virological response after early antiviral treatment of acute hepatitis C virus and HIV coinfection. Clin Infect Dis 49: 466–472.

77. SchröckE, duMS, VeldmanT, SchoellB, WienbergJ, et al. (1996) Multicolor spectral karyotyping of human chromosomes. Science 273: 494–497.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 9
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#