#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

An In-Depth Comparison of Latent HIV-1 Reactivation in Multiple Cell Model Systems and Resting CD4+ T Cells from Aviremic Patients


The possibility of HIV-1 eradication has been limited by the existence of latently infected cellular reservoirs. Studies to examine control of HIV latency and potential reactivation have been hindered by the small numbers of latently infected cells found in vivo. Major conceptual leaps have been facilitated by the use of latently infected T cell lines and primary cells. However, notable differences exist among cell model systems. Furthermore, screening efforts in specific cell models have identified drug candidates for “anti-latency” therapy, which often fail to reactivate HIV uniformly across different models. Therefore, the activity of a given drug candidate, demonstrated in a particular cellular model, cannot reliably predict its activity in other cell model systems or in infected patient cells, tested ex vivo. This situation represents a critical knowledge gap that adversely affects our ability to identify promising treatment compounds and hinders the advancement of drug testing into relevant animal models and clinical trials. To begin to understand the biological characteristics that are inherent to each HIV-1 latency model, we compared the response properties of five primary T cell models, four J-Lat cell models and those obtained with a viral outgrowth assay using patient-derived infected cells. A panel of thirteen stimuli that are known to reactivate HIV by defined mechanisms of action was selected and tested in parallel in all models. Our results indicate that no single in vitro cell model alone is able to capture accurately the ex vivo response characteristics of latently infected T cells from patients. Most cell models demonstrated that sensitivity to HIV reactivation was skewed toward or against specific drug classes. Protein kinase C agonists and PHA reactivated latent HIV uniformly across models, although drugs in most other classes did not.


Vyšlo v časopise: An In-Depth Comparison of Latent HIV-1 Reactivation in Multiple Cell Model Systems and Resting CD4+ T Cells from Aviremic Patients. PLoS Pathog 9(12): e32767. doi:10.1371/journal.ppat.1003834
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003834

Souhrn

The possibility of HIV-1 eradication has been limited by the existence of latently infected cellular reservoirs. Studies to examine control of HIV latency and potential reactivation have been hindered by the small numbers of latently infected cells found in vivo. Major conceptual leaps have been facilitated by the use of latently infected T cell lines and primary cells. However, notable differences exist among cell model systems. Furthermore, screening efforts in specific cell models have identified drug candidates for “anti-latency” therapy, which often fail to reactivate HIV uniformly across different models. Therefore, the activity of a given drug candidate, demonstrated in a particular cellular model, cannot reliably predict its activity in other cell model systems or in infected patient cells, tested ex vivo. This situation represents a critical knowledge gap that adversely affects our ability to identify promising treatment compounds and hinders the advancement of drug testing into relevant animal models and clinical trials. To begin to understand the biological characteristics that are inherent to each HIV-1 latency model, we compared the response properties of five primary T cell models, four J-Lat cell models and those obtained with a viral outgrowth assay using patient-derived infected cells. A panel of thirteen stimuli that are known to reactivate HIV by defined mechanisms of action was selected and tested in parallel in all models. Our results indicate that no single in vitro cell model alone is able to capture accurately the ex vivo response characteristics of latently infected T cells from patients. Most cell models demonstrated that sensitivity to HIV reactivation was skewed toward or against specific drug classes. Protein kinase C agonists and PHA reactivated latent HIV uniformly across models, although drugs in most other classes did not.


Zdroje

1. FinziD, HermankovaM, PiersonT, CarruthLM, BuckC, et al. (1997) Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 278: 1295–1300.

2. ChunTW, CarruthL, FinziD, ShenX, DiGiuseppeJA, et al. (1997) Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature 387: 183–188.

3. WongJK, HezarehM, GunthardHF, HavlirDV, IgnacioCC, et al. (1997) Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 278: 1291–1295.

4. DeeksSG (2012) HIV: Shock and kill. Nature 487: 439–440.

5. FolksTM, ClouseKA, JustementJ, RabsonA, DuhE, et al. (1989) Tumor necrosis factor alpha induces expression of human immunodeficiency virus in a chronically infected T-cell clone. Proc Natl Acad Sci U S A 86: 2365–2368.

6. PomerantzRJ, TronoD, FeinbergMB, BaltimoreD (1990) Cells nonproductively infected with HIV-1 exhibit an aberrant pattern of viral RNA expression: a molecular model for latency. Cell 61: 1271–1276.

7. AntoniBA, RabsonAB, KinterA, BodkinM, PoliG (1994) NF-kappa B-dependent and -independent pathways of HIV activation in a chronically infected T cell line. Virology 202: 684–694.

8. JordanA, BisgroveD, VerdinE (2003) HIV reproducibly establishes a latent infection after acute infection of T cells in vitro. Embo J 22: 1868–1877.

9. Micheva-VitevaS, KobayashiY, EdelsteinLC, PacchiaAL, LeeHL, et al. (2011) High-throughput screening uncovers a compound that activates latent HIV-1 and acts cooperatively with a histone deacetylase (HDAC) inhibitor. The Journal of biological chemistry 286: 21083–21091.

10. DuvergerA, JonesJ, MayJ, Bibollet-RucheF, WagnerFA, et al. (2009) Determinants of the establishment of human immunodeficiency virus type 1 latency. J Virol 83: 3078–3093.

11. KauderSE, BosqueA, LindqvistA, PlanellesV, VerdinE (2009) Epigenetic regulation of HIV-1 latency by cytosine methylation. PLoS Pathog 5: e1000495.

12. PaceMJ, AgostoL, GrafEH, O'DohertyU (2011) HIV reservoirs and latency models. Virology 411: 344–354.

13. HakreS, ChavezL, ShirakawaK, VerdinE (2012) HIV latency: experimental systems and molecular models. FEMS Microbiol Rev 36: 706–716.

14. BosqueA, PlanellesV (2011) Studies of HIV-1 latency in an ex vivo model that uses primary central memory T cells. Methods 53: 54–61.

15. BosqueA, PlanellesV (2009) Induction of HIV-1 latency and reactivation in primary memory CD4+ T cells. Blood 113: 58–65.

16. MariniA, HarperJM, RomerioF (2008) An in vitro system to model the establishment and reactivation of HIV-1 latency. J Immunol 181: 7713–7720.

17. YangHC, XingS, ShanL, O'ConnellK, DinosoJ, et al. (2009) Small-molecule screening using a human primary cell model of HIV latency identifies compounds that reverse latency without cellular activation. J Clin Invest 119 (11) 3473–86.

18. TyagiM, PearsonRJ, KarnJ (2010) Establishment of HIV latency in primary CD4+ cells is due to epigenetic transcriptional silencing and P-TEFb restriction. J Virol 84: 6425–6437.

19. ContrerasX, BarboricM, LenasiT, PeterlinBM (2007) HMBA releases P-TEFb from HEXIM1 and 7SK snRNA via PI3K/Akt and activates HIV transcription. PLoS Pathog 3: 1459–1469.

20. SwiggardWJ, BaytopC, YuJJ, DaiJ, LiC, et al. (2005) Human immunodeficiency virus type 1 can establish latent infection in resting CD4+ T cells in the absence of activating stimuli. J Virol 79: 14179–14188.

21. SalehS, SolomonA, WightmanF, XhilagaM, CameronPU, et al. (2007) CCR7 ligands CCL19 and CCL21 increase permissiveness of resting memory CD4+ T cells to HIV-1 infection: a novel model of HIV-1 latency. Blood 110: 4161–4164.

22. ArchinNM, EronJJ, PalmerS, Hartmann-DuffA, MartinsonJA, et al. (2008) Valproic acid without intensified antiviral therapy has limited impact on persistent HIV infection of resting CD4+ T cells. AIDS 22: 1131–1135.

23. LassenKG, HebbelerAM, BhattacharyyaD, LobritzMA, GreeneWC (2012) A flexible model of HIV-1 latency permitting evaluation of many primary CD4 T-cell reservoirs. PLoS One 7: e30176.

24. CameronPU, SalehS, SallmannG, SolomonA, WightmanF, et al. (2010) Establishment of HIV-1 latency in resting CD4+ T cells depends on chemokine-induced changes in the actin cytoskeleton. Proc Natl Acad Sci U S A 107: 16934–16939.

25. MessiM, GiacchettoI, NagataK, LanzavecchiaA, NatoliG, et al. (2003) Memory and flexibility of cytokine gene expression as separable properties of human T(H)1 and T(H)2 lymphocytes. Nat Immunol 4: 78–86.

26. SpinaCA, GuatelliJC, RichmanDD (1995) Establishment of a stable, inducible form of human immunodeficiency virus type 1 DNA in quiescent CD4 lymphocytes in vitro. J Virol 69: 2977–2988.

27. SpinaCA, PrinceHE, RichmanDD (1997) Preferential replication of HIV-1 in the CD45RO memory cell subset of primary CD4 lymphocytes in vitro. J Clin Invest 99: 1774–1785.

28. ChanJKL, BhattacharyyaD, LassenK, RuelasD, GreeneWC (2013) Calcium/Calcineurin Synergizes with Prostratin to Promote NF-kappa B Dependent Activation of Latent HIV. PLoS ONE 8 (10) e77749.

29. BlanksonJN, FinziD, PiersonTC, SabundayoBP, ChadwickK, et al. (2000) Biphasic decay of latently infected CD4+ T cells in acute human immunodeficiency virus type 1 infection. J Infect Dis 182: 1636–1642.

30. RisuenoRM, SchamelWW, AlarconB (2008) T cell receptor engagement triggers its CD3epsilon and CD3zeta subunits to adopt a compact, locked conformation. PLoS One 3: e1747.

31. LedbetterJA, ImbodenJB, SchievenGL, GrosmaireLS, RabinovitchPS, et al. (1990) CD28 ligation in T-cell activation: evidence for two signal transduction pathways. Blood 75: 1531–1539.

32. Mochly-RosenD, DasK, GrimesKV (2012) Protein kinase C, an elusive therapeutic target? Nat Rev Drug Discov 11: 937–957.

33. KulkoskyJ, CulnanDM, RomanJ, DornadulaG, SchnellM, et al. (2001) Prostratin: activation of latent HIV-1 expression suggests a potential inductive adjuvant therapy for HAART. Blood 98: 3006–3015.

34. GustafsonKR, CardellinaJH2nd, McMahonJB, GulakowskiRJ, IshitoyaJ, et al. (1992) A nonpromoting phorbol from the samoan medicinal plant Homalanthus nutans inhibits cell killing by HIV-1. J Med Chem 35: 1978–1986.

35. GulakowskiRJ, McMahonJB, BuckheitRWJr, GustafsonKR, BoydMR (1997) Antireplicative and anticytopathic activities of prostratin, a non-tumor-promoting phorbol ester, against human immunodeficiency virus (HIV). Antiviral Res 33: 87–97.

36. Trindade-SilvaAE, Lim-FongGE, SharpKH, HaygoodMG (2010) Bryostatins: biological context and biotechnological prospects. Curr Opin Biotechnol 21: 834–842.

37. MehlaR, Bivalkar-MehlaS, ZhangR, HandyI, AlbrechtH, et al. (2010) Bryostatin modulates latent HIV-1 infection via PKC and AMPK signaling but inhibits acute infection in a receptor independent manner. PLoS One 5: e11160.

38. DeChristopherBA, LoyBA, MarsdenMD, SchrierAJ, ZackJA, et al. (2012) Designed, synthetically accessible bryostatin analogues potently induce activation of latent HIV reservoirs in vitro. Nat Chem 4: 705–710.

39. LuznikL, KrausG, GuatelliJ, RichmanD, Wong-StaalF (1995) Tat-independent replication of human immunodeficiency viruses. J Clin Invest 95: 328–332.

40. WangFX, XuY, SullivanJ, SouderE, ArgyrisEG, et al. (2005) IL-7 is a potent and proviral strain-specific inducer of latent HIV-1 cellular reservoirs of infected individuals on virally suppressive HAART. J Clin Invest 115: 128–137.

41. LehrmanG, YlisastiguiL, BoschRJ, MargolisDM (2004) Interleukin-7 induces HIV type 1 outgrowth from peripheral resting CD4+ T cells. J Acquir Immune Defic Syndr 36: 1103–1104.

42. Scripture-AdamsDD, BrooksDG, KorinYD, ZackJA (2002) Interleukin-7 induces expression of latent human immunodeficiency virus type 1 with minimal effects on T-cell phenotype. J Virol 76: 13077–13082.

43. BosqueA, FamigliettiM, WeyrichAS, GoulstonC, PlanellesV (2011) Homeostatic proliferation fails to efficiently reactivate HIV-1 latently infected central memory CD4+ T cells. PLoS Pathog 7: e1002288.

44. SeddonB, TomlinsonP, ZamoyskaR (2003) Interleukin 7 and T cell receptor signals regulate homeostasis of CD4 memory cells. Nat Immunol 4: 680–686.

45. VandergeetenC, FromentinR, DaFonsecaS, LawaniMB, SeretiI, et al. (2013) Interleukin-7 promotes HIV persistence during antiretroviral therapy. Blood 121: 4321–4329.

46. SalehS, WightmanF, RamanayakeS, AlexanderM, KumarN, et al. (2011) Expression and reactivation of HIV in a chemokine induced model of HIV latency in primary resting CD4+ T cells. Retrovirology 8: 80.

47. GreeneWC, RobbRJ, DepperJM, LeonardWJ, DrogulaC, et al. (1984) Phorbol diester induces expression of Tac antigen on human acute T lymphocytic leukemic cells. J Immunol 133: 1042–1047.

48. RichardsonJH, SodroskiJG, WaldmannTA, MarascoWA (1995) Phenotypic knockout of the high-affinity human interleukin 2 receptor by intracellular single-chain antibodies against the alpha subunit of the receptor. Proc Natl Acad Sci U S A 92: 3137–3141.

49. KimHR, HwangKA, KimKC, KangI (2007) Down-regulation of IL-7Ralpha expression in human T cells via DNA methylation. J Immunol 178: 5473–5479.

50. DuhEJ, MauryWJ, FolksTM, FauciAS, RabsonAB (1989) Tumor necrosis factor alpha activates human immunodeficiency virus type 1 through induction of nuclear factor binding to the NF-kappa B sites in the long terminal repeat. Proc Natl Acad Sci U S A 86: 5974–5978.

51. OsbornL, KunkelS, NabelGJ (1989) Tumor necrosis factor alpha and interleukin 1 stimulate the human immunodeficiency virus enhancer by activation of the nuclear factor kappa B. Proc Natl Acad Sci U S A 86: 2336–2340.

52. SiegelDS, ZhangX, FeinmanR, TeitzT, ZelenetzA, et al. (1998) Hexamethylene bisacetamide induces programmed cell death (apoptosis) and down-regulates BCL-2 expression in human myeloma cells. Proc Natl Acad Sci U S A 95: 162–166.

53. RichonVM, WebbY, MergerR, SheppardT, JursicB, et al. (1996) Second generation hybrid polar compounds are potent inducers of transformed cell differentiation. Proc Natl Acad Sci U S A 93: 5705–5708.

54. VlachJ, PithaPM (1993) Hexamethylene bisacetamide activates the human immunodeficiency virus type 1 provirus by an NF-kappa B-independent mechanism. J Gen Virol 74 (Pt 11) 2401–2408.

55. ChenR, LiuM, LiH, XueY, RameyWN, et al. (2008) PP2B and PP1alpha cooperatively disrupt 7SK snRNP to release P-TEFb for transcription in response to Ca2+ signaling. Genes Dev 22: 1356–1368.

56. ChoudharySK, ArchinNM, MargolisDM (2008) Hexamethylbisacetamide and disruption of human immunodeficiency virus type 1 latency in CD4(+) T cells. J Infect Dis 197: 1162–1170.

57. TrujilloKM, TylerRK, YeC, BergerSL, OsleyMA (2011) A genetic and molecular toolbox for analyzing histone ubiquitylation and sumoylation in yeast. Methods 54: 296–303.

58. GardnerKE, AllisCD, StrahlBD (2011) Operating on chromatin, a colorful language where context matters. J Mol Biol 409: 36–46.

59. StruhlK (1998) Histone acetylation and transcriptional regulatory mechanisms. Genes Dev 12: 599–606.

60. ArchinNM, KeedyKS, EspesethA, DangH, HazudaDJ, et al. (2009) Expression of latent human immunodeficiency type 1 is induced by novel and selective histone deacetylase inhibitors. AIDS 23: 1799–1806.

61. KeedyKS, ArchinNM, GatesAT, EspesethA, HazudaDJ, et al. (2009) A limited group of class I histone deacetylases acts to repress human immunodeficiency virus type 1 expression. J Virol 83: 4749–4756.

62. ContrerasX, SchwenekerM, ChenCS, McCuneJM, DeeksSG, et al. (2009) Suberoylanilide Hydroxamic Acid Reactivates HIV from Latently Infected Cells. J Biol Chem 284: 6782–6789.

63. EdelsteinLC, Micheva-VitevaS, PhelanBD, DoughertyJP (2009) Short communication: activation of latent HIV type 1 gene expression by suberoylanilide hydroxamic acid (SAHA), an HDAC inhibitor approved for use to treat cutaneous T cell lymphoma. AIDS Res Hum Retroviruses 25: 883–887.

64. RasmussenTA, Schmeltz SogaardO, BrinkmannC, WightmanF, LewinSR, et al. (2013) Comparison of HDAC inhibitors in clinical development: Effect on HIV production in latently infected cells and T-cell activation. Hum Vaccin Immunother 9 9 (5) 993–1001.

65. ReuseS, CalaoM, KabeyaK, GuiguenA, GatotJS, et al. (2009) Synergistic activation of HIV-1 expression by deacetylase inhibitors and prostratin: implications for treatment of latent infection. PLoS One 4: e6093.

66. 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.

67. BouchatS, GatotJS, KabeyaK, CardonaC, ColinL, et al. (2012) Histone methyltransferase inhibitors induce HIV-1 recovery in resting CD4(+) T cells from HIV-1-infected HAART-treated patients. AIDS 26: 1473–1482.

68. BlazkovaJ, ChunTW, BelayBW, MurrayD, JustementJS, et al. (2012) Effect of histone deacetylase inhibitors on HIV production in latently infected, resting CD4(+) T cells from infected individuals receiving effective antiretroviral therapy. J Infect Dis 206: 765–769.

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

70. BartholomeeusenK, FujinagaK, XiangY, PeterlinBM (2013) HDAC inhibitors that release Positive Transcription Elongation Factor b (P-TEFb) from its Inhibitory Complex also activate HIV Transcription. J Biol Chem 288 (20) 14400–7.

71. PrinsJM, JurriaansS, van PraagRM, BlaakH, van RijR, et al. (1999) Immuno-activation with anti-CD3 and recombinant human IL-2 in HIV-1-infected patients on potent antiretroviral therapy. AIDS 13: 2405–2410.

72. van PraagRM, PrinsJM, RoosMT, SchellekensPT, Ten BergeIJ, et al. (2001) OKT3 and IL-2 treatment for purging of the latent HIV-1 reservoir in vivo results in selective long-lasting CD4+ T cell depletion. J Clin Immunol 21: 218–226.

73. BudhirajaS, FamigliettiM, BosqueA, PlanellesV, RiceAP (2012) Cyclin T1 and CDK9 T-loop phosphorylation are downregulated during establishment of HIV-1 latency in primary resting memory CD4+ T cells. J Virol 87 (2) 1211–20.

74. LenasiT, ContrerasX, PeterlinBM (2008) Transcriptional interference antagonizes proviral gene expression to promote HIV latency. Cell Host Microbe 4: 123–133.

75. IrieK, YanagitaRC, NakagawaY (2010) Challenges to the development of bryostatin-type anticancer drugs based on the activation mechanism of protein kinase Cdelta. Med Res Rev 32 (3) 518–35.

76. PerezM, de VinuesaAG, Sanchez-DuffhuesG, MarquezN, BellidoML, et al. (2010) Bryostatin-1 synergizes with histone deacetylase inhibitors to reactivate HIV-1 from latency. Curr HIV Res 8: 418–429.

77. SchroderAR, ShinnP, ChenH, BerryC, EckerJR, et al. (2002) HIV-1 integration in the human genome favors active genes and local hotspots. Cell 110: 521–529.

78. LewinskiMK, BisgroveD, ShinnP, ChenH, HoffmannC, et al. (2005) Genome-wide analysis of chromosomal features repressing human immunodeficiency virus transcription. J Virol 79: 6610–6619.

79. HanY, LinYB, AnW, XuJ, YangHC, et al. (2008) Orientation-dependent regulation of integrated HIV-1 expression by host gene transcriptional readthrough. Cell Host Microbe 4: 134–146.

80. ShanL, YangHC, RabiSA, BravoHC, ShroffNS, et al. (2011) Influence of host gene transcription level and orientation on HIV-1 latency in a primary-cell model. J Virol 85: 5384–5393.

81. Sherrill-MixS, LewinskiMK, FamigliettiM, BosqueA, MalaniN, et al. (2013) HIV latency and integration site placement in five cell-based models. Retrovirology 10: 90.

82. PaceMJ, GrafEH, AgostoLM, MexasAM, MaleF, et al. (2012) Directly infected resting CD4+T cells can produce HIV Gag without spreading infection in a model of HIV latency. PLoS Pathog 8: e1002818.

83. LewinSR, MurrayJM, SolomonA, WightmanF, CameronPU, et al. (2008) Virologic determinants of success after structured treatment interruptions of antiretrovirals in acute HIV-1 infection. J Acquir Immune Defic Syndr 47: 140–147.

84. KoelschKK, LiuL, HaubrichR, MayS, HavlirD, et al. (2008) Dynamics of total, linear nonintegrated, and integrated HIV-1 DNA in vivo and in vitro. J Infect Dis 197: 411–419.

85. MarksPA, BreslowR (2007) Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotechnol 25: 84–90.

86. de HoonMJ, ImotoS, NolanJ, MiyanoS (2004) Open source clustering software. Bioinformatics 20: 1453–1454.

87. SaldanhaAJ (2004) Java Treeview–extensible visualization of microarray data. Bioinformatics 20: 3246–3248.

88. SuzukiR, ShimodairaH (2006) Pvclust: an R package for assessing the uncertainty in hierarchical clustering. Bioinformatics 22: 1540–1542.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 12
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Eozinofilní granulomatóza s polyangiitidou
nový kurz
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#